Next Article in Journal
A Prospective Validation Study of Lung Cancer Gene Panel Testing Using Cytological Specimens
Next Article in Special Issue
Immunohistochemical Markers in Endometrial Cancer: Latest Updates
Previous Article in Journal
Tyrosine Kinase Inhibitors in the Treatment of Metastasised Renal Cell Carcinoma—Future or the Past?
Previous Article in Special Issue
Testing for Lynch Syndrome in Endometrial Carcinoma: From Universal to Age-Selective MLH1 Methylation Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

The Role of Immunohistochemistry Markers in Endometrial Cancer with Mismatch Repair Deficiency: A Systematic Review

1
Department of Gynecological and Breast Surgery and Oncology, Pitié-Salpêtrière, Assistance Publique des Hôpitaux de Paris (AP-HP), University Hospital, 75013 Paris, France
2
Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, INSERM, Sorbonne Université, 75012 Paris, France
3
Department of Pathology, Tenon Hospital, HUEP, Sorbonne Université, 75020 Paris, France
4
Centre de Recherche Saint-Antoine (CRSA), INSERM UMR_S_938, Cancer Biology and Therapeutics, Sorbonne University, 75012 Paris, France
5
Department of Pathology, Pitié-Salpêtrière Hospital, Sorbonne University, 75013 Paris, France
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(15), 3783; https://doi.org/10.3390/cancers14153783
Submission received: 5 June 2022 / Revised: 28 July 2022 / Accepted: 29 July 2022 / Published: 3 August 2022
(This article belongs to the Special Issue Immunohistochemical Markers in Endometrial Cancer)

Abstract

:

Simple Summary

Identification of mismatch repair-deficient tumors (MMRd), which occur in up to 30% of all endometrial cancers (EC), has become unavoidable for therapeutic management, clinical decision making, and prognosis. The objective of this systematic review was to summarize our current knowledge of the role of immunohistochemistry (IHC) markers for identifying MMRd tumors in EC. IHC with the expression of four proteins and MLH1 promoter methylation remains the reference of choice for diagnosis because it is reproducible and applicable in routine clinical practice. Further studies are needed to evaluate IHC in comparison with molecular tests including artificial intelligence, in terms of both efficacy and medical/economic aspects.

Abstract

The objective of this systematic review was to summarize our current knowledge of the role of immunohistochemistry (IHC) markers for identifying mismatch repair-deficient (MMRd) tumors in endometrial cancer (EC). Identification of MMRd tumors, which occur in 13% to 30% of all ECs, has become critical for patients with colorectal and endometrial cancer for therapeutic management, clinical decision making, and prognosis. This review was conducted by two authors applying the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines using the following terms: “immunohistochemistry and microsatellite instability endometrial cancer” or “immunohistochemistry and mismatch repair endometrial cancer” or “immunohistochemistry and mismatch repair deficient endometrial cancer”. Among 596 retrieved studies, 161 fulfilled the inclusion criteria. Articles were classified and presented according to their interest for the diagnosis, prognosis, and theragnostics for patients with MMRd EC. We identified 10, 18, and 96 articles using IHC expression of two, three, or four proteins of the MMR system (MLH1, MSH2, MHS6, and PMS2), respectively. MLH1 promoter methylation was analyzed in 57 articles. Thirty-four articles classified MMRd tumors with IHC markers according to their prognosis in terms of recurrence-free survival (RFS), overall survival (OS), stage, grade, and lymph node invasion. Theragnostics were studied in eight articles underlying the important concentration of PD-L1 in MMRd EC. Even though the role of IHC has been challenged, it represents the most common, robust, and cheapest method for diagnosing MMRd tumors in EC and is a valuable tool for exploring novel biotherapies and treatment modalities.

1. Introduction

In 2020, an estimated 417,367 new cases of endometrial cancer (EC) were diagnosed worldwide, and 97,370 women are estimated to have died from the disease [1]. Most ECs (90%) are diagnosed at an early stage with a 5 year survival rate of 95% [2]. However, once the cancer spreads to distant organs, the prognosis is poor with a 5 year survival rate of 17% [3].
In the past decade, numerous studies have explored prognostic factors such as pathologic type, histologic grade, lymphovascular involvement, and tumor staging but with insufficient reproducibility. Investigation has, therefore, turned to gene carcinogenesis such as molecular alterations to provide a new prognostic classification.
In 2017, the Proactive Molecular Risk Classifier for Endometrial Cancer (ProMisE) described four molecular prognostic groups: ultramutated DNA polymerase epsilon (POLE) tumors which have the best prognosis, microsatellite instability (MSI) or mismatch repair (MMR)-deficient (MMRd) hypermutated tumors with intermediate prognosis, p53 abnormal tumors with the worst prognosis, and tumors with copy-number low alterations with good to intermediate prognosis [4]. This classification is consistent with The Cancer Genome Atlas (TCGA) and applicable in clinical practice [5,6].
The MMRd tumor phenotype represents 17–33% of all ECs [7]. Originally, this molecular phenotype was found in patients with germinal mutations known as Lynch syndrome (LS) [8], which conveys a lifetime risk of EC of 60% [9]. Furthermore, this specific molecular genetic alteration, resulting from a defect in the MMR genes hMLH1, hMSH2, hMSH6, or hPMS2 is found in sporadic tumors called MMRd or Lynch-like syndrome tumors [10]. Tumors without defect in MMR genes are called mismatch repair-proficient (MMRp). The accumulation of insertions or deletions of nucleotides into coding repeat sequences results in an increase in lymphocyte infiltration, and the phenotype is, therefore, a possible candidate for immunotherapy [11]. Thus, identification of MMRd tumors has become critical for patients with EC for therapeutic management, clinical decision making, and prognosis.
In addition to MMR testing and to better identify risk groups currently included in the latest European Society of Gynecological Oncology/European Society for Radiotherapy and Oncology/European Society of Pathology (ESGO/ESTRO/ESP) [12], IHC markers such as p53 or POL-E have been proposed in a diagnostic algorithm [13]. Recent studies showed that abnormal p53 IHC reliably identifies cases with TP53 mutation (representing 25% of all EC) in EC biopsies (94% specificity and 91% sensitivity) [14]. POL-E mutations representing 8.59% of all ECs are mainly presented at earlier stages I–II (89.51%) and at the highest grade III (51.53%) [15]. Other IHC markers have been studied such as estrogen receptor, progesterone receptor, HER-2, and Ki67; however, no single marker was found to be indicative of EC often enough to allow routine use in the subclassification of EC [16].
Since 2018, the National Comprehensive Cancer Network guidelines recommend universal testing of all ECs for MSI/MMRd tumors [17]. Many oncologic centers use immunohistochemistry (IHC) for such testing as it is cheap and of high sensitivity, specificity, and reproducibility [5,18]. The European Society for Medical Oncology (ESMO) guidelines recommend the MMR-IHC test [12] for all patients with EC irrespective of histologic subtype. Molecular tests, such as PCR-based molecular testing using five of the eight mononucleotide or dinucleotide repeats (BAT-25, BAT-26, NR-21, NR-24, NR-27, D5S346, D2S123, and D17S250) or next-generation sequencing (NGS) can be used as an alternative or when IHC is indeterminate. Nevertheless, it is unclear which technique is recommended, reliable, and suitable for use in routine practice.
To the best of our knowledge, there is no recent review of the value of IHC markers to identify MMRd EC phenotypes or LS-related EC. The objective of this systematic review was, therefore, to summarize our current knowledge of the role of IHC markers in MMRd EC focusing on prognosis, diagnosis, and theragnostics.

2. Materials and Methods

This systematic review was carried out in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines using the following databases: MEDLINE, PubMed (the Internet portal of the National Library of Medicine, http://www.ncbi.nlm.nih.gov/sites/entrez?db=pubmed; accessed on 30 September 2021), the Cochrane Library, Cochrane databases “Cochrane Reviews”, and “Clinical Trials” (http://www3.interscience.wiley.com/cgi-bin/mrwhome/106568753/HOMEDARE; accessed on 30 September 2021). The PRISMA checklist is provide in the Supplementary Materials (Figure S1). This systematic review has not been registered in PROSPERO.
Two independent reviewers (A.F. and G.C.) retrieved information about articles such as authors, date of publication, journal, study design, population characteristics, type of screening test, number of MMRd tumors studied, outcomes, and survival rates. Any discrepancies about the eligibility of an article were discussed between the two reviewers, with the senior author (G.C.) having the final word.
The search was conducted with the following terms: “immunohistochemistry and microsatellite instability endometrial cancer” or “immunohistochemistry and mismatch repair endometrial cancer” or “immunohistochemistry and mismatch repair deficient endometrial cancer”. Lynch syndrome patients were also included.
The database search was further supplemented with original articles, reviews, and meta-analyses. All duplicates were removed. Only articles published in English between 1 July 1999 and 30 September 2021 were included. Among 596 retrieved studies, 161 fulfilled the inclusion criteria. The exclusion criteria were no detail of IHC, no MMRd tumors, no detail of EC, or MMRd diagnosed with molecular approach only.
The articles were classified and presented according to the use of IHC for the diagnosis, prognosis, and theragnostics for patients with MMRd endometrial tumors. We summarized the results of the main outcomes, as it was not feasible to pool findings due to the heterogeneity among the studies in terms of patients, tumor characteristics, technique used, statistical analysis, and outcome measures.

3. Results

The PRISMA flow diagram is presented in Figure 1.
This systematic review identified 157 original articles and four literature reviews reporting the role of IHC markers in MMRd EC. Of the 155 original articles studying neoplastic endometrial tissue, the average number of endometrial tumor samples was 389 (minimum four, maximum 5917). A total of 103 articles reported analysis from formalin-fixed paraffin-embedded (FFPE) tissue, along with eight from FFPE and frozen tissue, 10 from frozen tissue, and one from fresh tissue.

3.1. IHC in the Diagnosis of MMRd Tumors in EC

IHC expression of two proteins of the MMR system in EC is presented in Table 1.
We identified 10 articles (published between 1999 and 2020) using IHC expression of two proteins of the MMR system in EC, nine of which reported the loss of the MLH1 and MSH2 proteins, and one of which reported the loss of MSH6 and PMS2 [19,20,21,22,23,24,25,26,27,28]. Of the 10 studies, nine used FFPE tissue and two used frozen tissue, including one article comparing neoplastic endometrial tissue with the surrounding healthy tissue. A germline mutation was screened in four articles [21,22,25].
The average number of MMRd tumors included was 17.9 (minimum six, maximum 53). The percentage of MMRd tumors in the cohorts ranged from 13.5 to 100%. The loss of the MLH1 and MSH2 proteins in the MMRd tumors ranged from 12 to 83.3% and from 1.1 to 86%, respectively. The loss of the MSH6 and PMS2 proteins was reported at 0% and 100%, respectively. One article reported MLH1 promoter methylation in 77% of the MMRd tumors.
IHC expression of three proteins of the MMR system in EC is presented in Table 2.
We identified 18 articles (published between 2002 and 2020) using IHC expression of three proteins of the MMR system (MLH1, MSH2, and MSH6) in EC, of which 15 used FFPE tissues, and three used frozen tissue [29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46]. Two articles did not describe the technique used, and five articles compared neoplastic endometrial tissue with the surrounding healthy tissue.
A germline mutation was screened in 11 of the articles [29,31,33,34,35,36,39,40,42,44,46]. The average number of MMRd tumors included was 34.6 (minimum six, maximum 164). The percentage of MMRd tumors in the cohorts ranged from 20% to 100%. The loss of the MSH2, MLH1, and MSH6 proteins in the MMRd tumors ranged from 0% to 55.2%, from 0% to 100%, and from 0% to 66.7%, respectively. Four articles reported MLH1 promoter methylation in 14% to 70% of the MMRd tumors.
IHC expression of four proteins of the MMR system in EC is presented in Table 3.
We identified 96 articles (published between 2008 and 2021) using IHC expression of four proteins of the MMR system (MLH1, MSH2, MSH6, and PMS2) in EC, of which 73 used FFPE tissues, three used frozen tissue, and one used tumor cells [40,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142]. Twenty-one articles did not describe the technique, and eight compared neoplastic endometrial tissue with surrounding healthy tissue.
A germline mutation was screened in 53 of the articles [49,50,51,53,54,56,57,61,62,63,64,68,69,70,71,72,74,75,79,82,83,84,85,86,87,88,89,90,92,93,94,96,97,104,106,110,112,113,114,116,119,121,122,123,124,127,128,129,133,136,140]. The average number of MMRd tumors included was 98.5 (minimum one, maximum 2563). The percentage of MMRd tumors in the cohorts ranged from 5% to 100%. The loss of the MSH2, MLH1, MSH6, and PMS2 proteins in the MMRd tumors ranged from 0% to 75%, from 0% to 100%, from 0% to 80%, and from 2% to 100%, respectively. Twenty-seven articles reported MLH1 promoter methylation in 14% to 100% of the MMRd tumors.
MLH1 promoter methylation analysis in MMRd EC is presented in Table 4.
We identified 57 articles analyzing MLH1 promoter methylation [19,26,32,33,34,36,40,42,46,47,52,55,57,58,59,69,71,75,79,81,82,83,84,85,86,90,91,94,95,96,99,100,112,113,116,122,124,125,127,128,129,136,137,139,140,141,142,143,144,145,146,147,148,149,150,151,152]. Among these, 39 articles used bisulfite conversion of tumor DNA with PCR amplification, five used a methylation-specific multiplex ligation dependent probe-amplification technique, two used NGS, and one used a pyrosequencing technique. The MLH1 promoter methylation gene was found in 14% to 100% of the MMRd tumors. It was tested in order to identify a germline or somatic mutation.

3.2. IHC in the Prognosis of MMRd Tumors in EC

We identified 34 articles studying the prognosis of patients with MMRd tumors in EC [25,26,51,54,60,63,70,72,75,76,84,89,92,99,102,105,106,115,120,126,130,131,132,133,135,140,141,153,154,155,156,157,158,159]. The studies included an average of 191.5 samples of MMRd tumors (minimum 12, maximum 892).

3.2.1. Survival

The recurrence-free survival (RFS) and overall survival (OS) of patients with MMRd tumors in EC compared with patients with MMRp tumors are presented in Table 5.
MMRd tumors were associated with better RFS than MMRp tumors in four articles [75,99,102,158]. One reported a recurrence of 10% in patients with MMRd tumors versus 42% in those with MMRp tumors [99] with a median follow-up of 31 months for both MMR groups. Another reported a hazard ratio (HR) of 0.61 [102]. A third one reported a 5 year RFS of 95% or 87.9% versus 80.4% for MMRp tumors, 93% for POLE tumors, 52% for no specific molecular profile tumors, and 42% for p53 tumors [75]. The last study reported 36 months of RFS versus 9 months for MMRp tumors [158]. Two articles reported a worse RFS for patients with MMRd tumors than for those with MMRp tumors [54,120]; the 5-year RFS for MMRd tumors was 66% in the first and 71.1% in the second compared with 89% and 97.6% for MMRp tumors.
MMRd tumors were associated with better OS than MMRp tumors in two articles [99,102]. With a median follow-up of 31 months, the mortality rate of patients with MMRd tumors was 13.1% compared to 36.1% for MMRp tumors (HR: 0.80). Two articles found a worse OS for patients with MMRd tumors with a 5 year OS of 74% and 71% compared with 86% and 100% for MMRp tumors [54,120]. Two articles showed an intermediate prognosis for MMRd tumors [126,131]. The 5 year RFS and OS for MMRd tumors were 71.4% and 81.3% compared with 98% and 98% for POLE tumors and 48% and 54% for p53 tumors, respectively. Lastly, in nine articles, there were no significant differences in the RFS and OS or clinical features between patients with MMRd or MMRp tumors [60,63,76,133,140,141,153,154,155].

3.2.2. Pathologic Characteristics

MMRd tumors were associated with a lower grade EC in three articles [106,115,156]; 83% to 90% of low-grade EC tumors had the MMRd phenotype compared with 31.4% to 77% of the MMRp tumor phenotypes. Five articles showed that MMRd tumors were associated with a higher-grade EC [25,70,130,132,157]; 33% of higher-grade tumors were found to be MMRd and associated with endometrioid and mixed histologic tumors.
MMRd tumors were associated with a lower FIGO stage in three articles [92,106,115]; 58% to 90% of low-stage EC were MMRd compared with 41.8% to 78% for MMRp tumors. MMRd tumors were associated with higher FIGO stage in six articles [25,72,132,135,157,160]; 58% to 66.7% in higher-stage EC were MMRd compared with 17.6% to 40.6% for MMRp tumors.
MMRd tumors were associated with less lymph node invasion in one article [157], with 44.6% compared with 55.4% in MMRp tumors.
MMRd tumors were associated with greater lymphovascular invasion in one article [92], with 58% for MMRd tumors and 48% for MMRp tumors.
In three articles MMRd tumors were associated with a higher probability of metastasis than MMRp tumors [26,72,89]; 75% of patients with metastasis had MMRd tumors compared with 11.5% of patients with MMRp tumors (OR: 7.44).

3.3. IHC in the Theragnostics of MMRd Tumors in EC

We identified eight articles which investigated the role of IHC in the theragnostics for MMRd tumors in EC [53,88,93,95,122,136,161,162].
The studies included an average of 60.3 samples of MMRd tumors (minimum two, maximum 203).
PD-L1 expression is presented in Table 6.
Immune checkpoint pathways such as PD-1/PD-L1 are the main target for immunotherapy in EC. We identified four articles studying PD-L1 expression in MMRd tumors [88,95,136,161]. All the articles reported an increase in the expression of PD-L1 ranging from 60.4% to 100% in the peritumoral or tumoral compartment in MMRd tumors. Only 5.3% of MMRp tumors were reported to have PD-L1 expression.
One article reported a study of adjuvant radiotherapy in non-endometrioid MMRd tumors and showed a progression-free survival (PFS) and OS of 5 years compared with a PFS and OS of 2 years for non-endometrioid MMRp tumors [53].
We identified one article showing that MMRd tumors were associated with a significantly lower mean percentage of androgen receptor (AR) expression [122]; AR expression was 65.9% in MMRd tumors compared with 81.6% in MMRp tumors.
Lastly, one article showed that MMRp tumors were more sensitive to progestin treatment; no patients with MMRd tumors showed disease regression compared with 41% of patient with MMRp tumors [93].

4. Discussion

This is the first systematic review of the value of IHC markers in EC with MMRd tumors. IHC with expression of all four proteins and MLH1 promoter methylation remains the reference of choice for diagnosis because it is reproducible and applicable in routine clinical practice. The search for MMRd tumors is becoming essential for the care management of patients with EC regarding the diagnostic, prognostic, and theragnostic evaluations. We identified 10 articles using IHC expression of two proteins of the MMR repair system in EC with a detection rate from 13.5% to 100%, 18 articles using IHC expression of three proteins with a detection rate from 20% to 100%, and 96 articles using IHC expression of four proteins with a detection rate from 5% to 100%. Fifty-seven articles analyzed MLH1 promoter methylation with a detection rate from 14% to 100%. Overall, most of the articles suggest a better prognosis for MMRd tumors versus MMRp tumors. After a median follow-up of 31 months (1–99 months), there was no difference in progression or recurrence rates between pMMR and dMMR tumors (19.5% vs. 16.5%; p = 0.31). However, among those with non-endometrioid tumors, recurrence and mortality rates were significantly higher for pMMR than dMMR tumors (42.0% vs. 10.0%, p = 0.001, and 36.1% vs. 13.1%, p = 0.01, respectively), despite similar stage and lymphovascular space invasion distributions. Lastly, in the four articles studying PD-L1, all reported increased expression in MMRd tumors ranging from 60.4% to 100%.
In the early 1990s, and in the context of hereditary nonpolyposis colorectal cancer (HNPCC) development, the main focus was on mutations in MLH1 and MSH2 genes [163]. It was first thought that the four MMR proteins functioned as a heterodimer complex only; in the absence of one of the two proteins (MLH1 with PMS2 and MSH2 with MSH6), the complex no longer functions and the other protein is not expressed [164,165,166]. This explains why oncology departments initially routinely used two proteins alone in IHC analysis and not four. In our review, articles analyzing the expression of two proteins included an average of 17.9 MMRd tumors, whereas articles analyzing the expression of three and four proteins included an average of 34.6 and 98.5 MMRd tumors, respectively. Thus, the use of four proteins in IHC increases the number of MMRd cases identified. In addition, Goodfellow et al., in a cohort of 1002 patients with EC, showed that the most common MMR defect was MLH1 loss followed by combined MSH2/MSH6 losses, then MSH6 loss alone at 70%, 20.5%, and 19.6%, respectively [71]. Several articles highlighted an increased prevalence of MSH6 mutations representing 3.8% (95% CI 1.0–9.5%) of patients with EC compared to 2.6% (95% CI 0.5–7.4%) of patients with HNPCC tumors [167,168,169]. It is, thus, essential to screen patients with EC with IHC using the four proteins. Screening with only two proteins would not only underestimate the prevalence of MMRd tumors in a cohort but would also misclassify patients as having MMRp tumors which would result in mistreatment.
As mentioned above, since 2018, the National Comprehensive Cancer Network recommends universal MMR testing for all newly diagnosed cases of EC [170]. Indeed, Mills et al. reported that 57.1% of patients with LS would not be identified on the basis of age and individual cancer history alone, and that 28.6% would not be identified even with a complete family history [82], implying that the Bethesda criteria are insufficient [171,172]. IHC has since become the standard practice in many institutions and is recommended in new guidelines as the gold standard screening test. As a consequence, recent articles studying the cost effectiveness of reflex testing for LS-related EC in patients older than 70 years that do not meet the Bethesda criteria suggest that the most cost-effective approach is to test all EC patients up to an age threshold (somewhere between 60 and 65 years) and that even testing all patients up to 70 or 80 years would be cost-effective compared with no testing. Using a cost-effectiveness threshold of 20,000 GBP per quality-adjusted-life-year (QALY), reflex testing for LS using MMR IHC and MLH1 methylation testing was cost-effective versus no testing, costing 14,200 GBP per QALY gained [173].
However, several other techniques, including molecular biology, are increasingly being studied. In our systematic review, we identified 96 studies including both IHC and MSI molecular analysis. Among these, 87 used a PCR-based molecular testing using five of the eight mononucleotide or dinucleotide repeats (BAT-25, BAT-26, NR-21, NR-24, NR-27, D5S346, D2S123, and D17S250), 15 used NGS, and two used the Idylla MSI test [174,175]. Sixteen articles reported MSI-High tumors when more than two markers were unstable, and two articles reported MSI-High when more than three markers were unstable. However, researchers have found other techniques to overcome the drawbacks of IHC. Stello et al., in large, randomized cohort trials in EC such as PORTEC-1 and PORTEC-2, concluded that MSI and IHC analyses are highly concordant (94%) [75]. In the same way, Stinton et al. showed no statistically significant differences in test accuracy estimates (sensitivity and specificity); the sensitivity of IHC ranged from 60.7–100%, and the sensitivity of MSI-based testing ranged from 41.7–100% [137]. Therefore, IHC remains the cheapest and most accurate test with a low failure rate to determine MMR. However, it would be interesting to find a new technique for the detection of MMRd tumors using artificial intelligence and molecular biology that is more manageable, faster, more reproducible, and less expensive than IHC.
IHC is essential to determine the prognosis for patients with certain tumors. In 2021, the ESMO described prognostic risks groups according to the stage, grade, lymphovascular involvement, myometrial invasion, and the molecular classification obtained by IHC to establish clinical and therapeutic guidelines [12].
In our systematic review, we identified 34 articles studying the prognosis in MMRd/MMRp tumors. A larger number of articles demonstrated a better RFS and a better OS for patients with MMRd tumors versus MMRp tumors, but these articles were based on a small number of patients. The article by Pina et al., consisting of 242 MMRd tumors and representing the largest MMRd cohort studying prognosis in our systematic review, showed that, among non-endometrioid tumors, recurrence and mortality rates were significantly higher for pMMR than dMMR tumors (42.0% vs. 10.0%, p = 0.001, and 36.1% vs. 13.1%, p = 0.01, respectively), despite similar stage and lymphovascular space invasion distributions [99]. We also identified controversial findings regarding the age at diagnosis, the stage, and the lymph node status of MMRd tumors. Studying 212 MMRd tumors, Tangjitgamol et al. found a significantly higher rate of MMRd tumors in patients aged less than 60 years, with early-stage disease, and more negative lymph node status than the other comparative groups: 59.2% vs. 48.3% (p = 0.037) for age, 58.2% vs. 45.2% (p = 0.027) for stage, and 58.1% vs. 44.6% (p = 0.048) for nodal status [92]. Despite studies to the contrary, new studies tend to describe MMRd tumors as tumors with a good prognosis compared to MMRp tumors. Knowledge of the MMR status is, therefore, essential for the therapeutic management of patients with EC.
Theragnostic studies are currently fashionable, and theragnostics are being increasingly used in patient management. In our systematic review, we identified eight articles studying the theragnostics of MMRd tumors in EC. All showed an increased expression, of up to 100%, of PD-L1 tumoral compartment in MMRd tumors compared to 5.3% in MMRp tumors. Currently, it is well established that PD-1 and PD-L1 checkpoints are the main target in immunotherapy for recurrence or refractory cases in several cancers [176,177]. Compared to other gynecological cancers, EC shows a large number of immune cells and intra-tumoral cytokines, which stimulate an endogenous antitumor immune response [178,179]. In a phase Ib/II clinical trial including patients with advanced EC receiving lenvatinib (a tyrosine kinase inhibitor) plus pembrolizumab, the objective response rate was 63.9% (95% CI: 30.8–80.9%) for MMRd tumors compared with 37.2% (95% CI: 37.5–47.8%) for MMRp tumors [180]. Immunotherapy for patients with EC, especially in cases of advanced or metastatic disease with MMRd tumors, is attracting considerable attention as more than 50 clinical trials investigating immunotherapy in EC have been listed on the clinicaltrials.gov website [181]. To date, three monoclonal antibodies targeting PD-1 (pembrolizumab, nivolumab, and cemiplimab) and three monoclonal antibodies targeting PD-L1 (atezolizumab, durvalumab, and avelumab) have been approved by the US Food and Drug Administration for advanced inoperable cancers in first-line, metastatic, and recurrent EC.
Our systematic review confirms Jumaah et al.’s meta-analysis which showed a large variation in the diagnosis of MMRds tumors. This depends on the initial study population, which may include only low- or high-grade tumors, Lynch families, or concurrent cancers. In contrast to us, this meta-analysis included MMRd tumors diagnosed by molecular biology, which probably allowed them to obtain a larger variation. From a prognostic point of view, our articles are concordant since they point out that MMRd tumors have a more favorable prognosis compared with MMR-proficient tumors, and that their immune context is a key point of immunotherapy [182].
This systematic review had some specific limitations. First, many studies did not detail their method of analysis for IHC, and the cohorts were heterogeneous, which made it impossible to synthesize the results in the form of a meta-analysis. The second limitation was the difficulty of comparing diagnostic techniques in terms of therapeutic effectiveness and cost-effectiveness. The strengths of our study were that the methodology used with the PRISMA Guidelines, and we included more than 161 articles classified by diagnosis, prognosis, and theragnostics. This review of the literature allows us to state that, in order not to underestimate the MSI tumors in the EC, it is strongly recommended to use the four proteins of the MMR system through the IHC technique, which remains the least expensive and most reproducible. Furthermore, our review updates the data in the literature on the prognosis of these tumors. The research on MSI tumors tends toward a better prognosis thanks to their particular immune context, which is key to many immunotherapies.

5. Conclusions

In this systematic review, we provided an overview of MMR status through IHC in EC. IHC with expression of all four proteins and MLH1 promoter methylation remains the reference of choice for diagnosis because it is reproducible, cheaper, and applicable in routine clinical practice. Molecular classification such as MMRd tumors has been essential to determine the prognosis. More and more clinical trials are using the immune context of MMRd tumors in immunotherapy treatment. Further studies are needed to evaluate IHC in comparison with molecular tests including artificial intelligence, in terms of both efficacy and medical/economic aspects.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/cancers14153783/s1. Figure S1: PRISMA checklist [183].

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics, 2022. CA A Cancer J. Clin. 2022, 72, 7–33. [Google Scholar] [CrossRef] [PubMed]
  3. Statistics Adapted from the American Cancer Society’s (ACS) Publication, Cancer Facts & Figures 2022, the ACS Website, and the International Agency for Research on Cancer Website. Available online: https://www.cancer.org/research/cancer-facts-statistics/all-cancer-facts-figures/cancer-facts-figures-2022.html (accessed on 1 January 2022).
  4. Talhouk, A.; McConechy, M.K.; Leung, S.; Yang, W.; Lum, A.; Senz, J.; Boyd, N.; Pike, J.; Anglesio, M.; Kwon, J.S.; et al. Confirmation of ProMisE: A simple, genomics-based clinical classifier for endometrial cancer: Molecular Classification of EC. Cancer 2017, 123, 802–813. [Google Scholar] [CrossRef] [Green Version]
  5. Travaglino, A.; Raffone, A.; Mascolo, M.; Guida, M.; Insabato, L.; Zannoni, G.F.; Zullo, F. Clear cell endometrial carcinoma and the TCGA classification. Histopathology 2020, 76, 336–368. [Google Scholar] [CrossRef] [PubMed]
  6. The Cancer Genome Atlas Research Network; Levine, D. Integrated genomic characterization of endometrial carcinoma. Nature 2013, 497, 67–73. [Google Scholar] [CrossRef]
  7. Colle, R.; Cohen, R. Épidémiologie des tumeurs MSI: Fréquence des tumeurs MSI en fonction de la localisation du cancer et de son stade. Bull. Cancer 2019, 106, 114–118. [Google Scholar] [CrossRef] [PubMed]
  8. Meyer, L.A.; Broaddus, R.R.; Lu, K.H. Endometrial Cancer and Lynch Syndrome: Clinical and Pathologic Considerations. Cancer Control. 2009, 16, 14–22. [Google Scholar] [CrossRef] [Green Version]
  9. Vasen, H.; Wijnen, J.; Menko, F.; Kleibeuker, J.; Taal, B.; Griffioen, G.; Nagengast, F.; Meijers-Heijboer, E.; Bertario, L.; Varesco, L.; et al. Cancer risk in families with hereditary nonpolyposis colorectal cancer diagnosed by mutation analysis. Gastroenterology 1996, 110, 1020–1027. [Google Scholar] [CrossRef]
  10. Fishel, R.; Kolodner, R.D. Identification of mismatch repair genes and their role in the development of cancer. Curr. Opin. Genet. Dev. 1995, 5, 382–395. [Google Scholar] [CrossRef]
  11. Ono, R.; Nakayama, K.; Nakamura, K.; Yamashita, H.; Ishibashi, T.; Ishikawa, M.; Minamoto, T.; Razia, S.; Ishikawa, N.; Otsuki, Y.; et al. Dedifferentiated Endometrial Carcinoma Could be A Target for Immune Checkpoint Inhibitors (Anti PD-1/PD-L1 Antibodies). IJMS 2019, 20, 3744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Concin, N.; Matias-Guiu, X.; Vergote, I.; Cibula, D.; Mirza, M.R.; Marnitz, S.; Ledermann, J.; Bosse, T.; Chargari, C.; Fagotti, A.; et al. ESGO/ESTRO/ESP guidelines for the management of patients with endometrial carcinoma. Int. J. Gynecol. Cancer 2021, 31, 12–39. [Google Scholar] [CrossRef] [PubMed]
  13. Vermij, L.; Smit, V.; Nout, R.; Bosse, T. Incorporation of molecular characteristics into endometrial cancer management. Histopathology 2020, 76, 52–63. [Google Scholar] [CrossRef] [PubMed]
  14. Singh, N.; Piskorz, A.M.; Bosse, T.; Jimenez-Linan, M.; Rous, B.; Brenton, J.D.; Gilks, C.B.; Köbel, M. p53 immunohistochemistry is an accurate surrogate for TP53 mutational analysis in endometrial carcinoma biopsies. J. Pathol. 2020, 250, 336–345. [Google Scholar] [CrossRef] [PubMed]
  15. Jumaah, A.S.; Salim, M.M.; Al-Haddad, H.S.; McAllister, K.A.; Yasseen, A.A. The frequency of POLE-mutation in endometrial carcinoma and prognostic implications: A systemic review and meta-analysis. J. Pathol. Transl. Med. 2020, 54, 471–479. [Google Scholar] [CrossRef] [PubMed]
  16. Li, M.; Zhao, L.; Qi, W.; Shen, D.; Li, X.; Wang, J.; Wei, L. Clinical implications and prognostic value of five biomarkers in endometrial carcinoma. Chin.-Ger J. Clin. Oncol. 2013, 12, 586–591. [Google Scholar] [CrossRef]
  17. Koh, W.-J.; Abu-Rustum, N.R.; Bean, S.; Bradley, K.; Campos, S.M.; Cho, K.R.; Chon, H.S.; Chu, C.; Cohn, D.; Crispens, M.A.; et al. Uterine Neoplasms, Version 1.2018, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Canc. Netw. 2018, 16, 170–199. [Google Scholar] [CrossRef] [Green Version]
  18. Raffone, A.; Travaglino, A.; Cerbone, M.; Gencarelli, A.; Mollo, A.; Insabato, L.; Zullo, F. Diagnostic Accuracy of Immunohistochemistry for Mismatch Repair Proteins as Surrogate of Microsatellite Instability Molecular Testing in Endometrial Cancer. Pathol. Oncol. Res. 2020, 26, 1417–1427. [Google Scholar] [CrossRef]
  19. Simpkins, S.B.; Bocker, T.; Swisher, E.M.; Mutch, D.G.; Gersell, D.J.; Kovatich, A.J.; Palazzo, J.P.; Fishel, R.; Goodfellow, P.J. MLH1 promoter methylation and gene silencing is the primary cause of microsatellite instability in sporadic endometrial cancers. Hum. Mol. Genet. 1999, 8, 661–666. [Google Scholar] [CrossRef] [Green Version]
  20. Peiró, G.; Diebold, J.; Mayr, D.; Baretton, G.B.; Kimmig, R.; Schmidt, M.; Löhrs, U. Prognostic relevance of hMLH1, hMSH2, and BAX protein expression in endometrial carcinoma. Mod. Pathol. 2001, 14, 777–783. [Google Scholar] [CrossRef]
  21. Maruyama, A.; Miyamoto, S.; Saito, T.; Kondo, H.; Baba, H.; Tsukamoto, N. Clinicopathologic and familial characteristics of endometrial carcinoma with multiple primary carcinomas in relation to the loss of protein expression of MSH2 and MLH1. Cancer 2001, 91, 2056–2064. [Google Scholar] [CrossRef]
  22. Berends, M.J.; Hollema, H.; Wu, Y.; van Der Sluis, T.; Mensink, R.G.; ten Hoor, K.A.; Sijmons, R.H.; de Vries, E.G.; Pras, E.; Mourits, M.J.; et al. MLH1 and MSH2 protein expression as a pre-screening marker in hereditary and non-hereditary endometrial hyperplasia and cancer. Int J. Cancer 2001, 92, 398–403. [Google Scholar] [CrossRef] [PubMed]
  23. Chiaravalli, A.M.; Furlan, D.; Facco, C.; Tibiletti, M.G.; Dionigi, A.; Casati, B.; Albarello, L.; Riva, C.; Capella, C. Immunohistochemical pattern of hMSH2/hMLH1 in familial and sporadic colorectal, gastric, endometrial and ovarian carcinomas with instability in microsatellite sequences. Virchows Arch. 2001, 438, 39–48. [Google Scholar] [CrossRef] [PubMed]
  24. Hardisson, D.; Moreno-Bueno, G.; Sánchez, L.; Sarrió, D.; Suárez, A.; Calero, F.; Palacios, J. Tissue microarray immunohistochemical expression analysis of mismatch repair (hMLH1 and hMSH2 genes) in endometrial carcinoma and atypical endometrial hyperplasia: Relationship with microsatellite instability. Mod. Pathol. 2003, 16, 1148–1158. [Google Scholar] [CrossRef] [Green Version]
  25. Sutter, C.; Dallenbach-Hellweg, G.; Schmidt, D.; Baehring, J.; Bielau, S.; von Knebel Doeberitz, M.; Gebert, J. Molecular analysis of endometrial hyperplasia in HNPCC-suspicious patients may predict progression to endometrial carcinoma. Int J. Gynecol. Pathol. 2004, 23, 18–25. [Google Scholar] [CrossRef]
  26. Irving, J.A.; Catasús, L.; Gallardo, A.; Bussaglia, E.; Romero, M.; Matias-Guiu, X.; Prat, J. Synchronous endometrioid carcinomas of the uterine corpus and ovary: Alterations in the beta-catenin (CTNNB1) pathway are associated with independent primary tumors and favorable prognosis. Hum. Pathol. 2005, 36, 605–619. [Google Scholar] [CrossRef]
  27. Alvarez, T.; Miller, E.; Duska, L.; Oliva, E. Molecular profile of grade 3 endometrioid endometrial carcinoma: Is it a type I or type II endometrial carcinoma? Am. J. Surg. Pathol. 2012, 36, 753–761. [Google Scholar] [CrossRef] [PubMed]
  28. Plotkin, A.; Kuzeljevic, B.; De Villa, V.; Thompson, E.F.; Gilks, C.B.; Clarke, B.A.; Köbel, M.; McAlpine, J.N. Interlaboratory Concordance of ProMisE Molecular Classification of Endometrial Carcinoma Based on Endometrial Biopsy Specimens. Int. J. Gynecol. Pathol. 2020, 39, 537–545. [Google Scholar] [CrossRef] [PubMed]
  29. Planck, M.; Rambech, E.; Möslein, G.; Müller, W.; Olsson, H.; Nilbert, M. High frequency of microsatellite instability and loss of mismatch-repair protein expression in patients with double primary tumors of the endometrium and colorectum. Cancer 2002, 94, 2502–2510. [Google Scholar] [CrossRef]
  30. Orbo, A.; Nilsen, M.N.; Arnes, M.S.; Pettersen, I.; Larsen, K. Loss of expression of MLH1, MSH2, MSH6, and PTEN related to endometrial cancer in 68 patients with endometrial hyperplasia. Int. J. Gynecol. Pathol. 2003, 22, 141–148. [Google Scholar] [CrossRef]
  31. Lipton, L.R.; Johnson, V.; Cummings, C.; Fisher, S.; Risby, P.; Eftekhar Sadat, A.T.; Cranston, T.; Izatt, L.; Sasieni, P.; Hodgson, S.V.; et al. Refining the Amsterdam Criteria and Bethesda Guidelines: Testing algorithms for the prediction of mismatch repair mutation status in the familial cancer clinic. J. Clin. Oncol. 2004, 22, 4934–4943. [Google Scholar] [CrossRef]
  32. Macdonald, N.D.; Salvesen, H.B.; Ryan, A.; Malatos, S.; Stefansson, I.; Iversen, O.E.; Akslen, L.A.; Das, S.; Jacobs, I.J. Molecular differences between RER+ and RER- sporadic endometrial carcinomas in a large population-based series. Int. J. Gynecol. Cancer 2004, 14, 957–965. [Google Scholar] [CrossRef] [PubMed]
  33. Buttin, B.M.; Powell, M.A.; Mutch, D.G.; Rader, J.S.; Herzog, T.J.; Gibb, R.K.; Huettner, P.; Edmonston, T.B.; Goodfellow, P.J. Increased risk for hereditary nonpolyposis colorectal cancer-associated synchronous and metachronous malignancies in patients with microsatellite instability-positive endometrial carcinoma lacking MLH1 promoter methylation. Clin. Cancer Res. 2004, 10, 481–490. [Google Scholar] [CrossRef] [Green Version]
  34. Soliman, P.T.; Broaddus, R.R.; Schmeler, K.M.; Daniels, M.S.; Gonzalez, D.; Slomovitz, B.M.; Gershenson, D.M.; Lu, K.H. Women with synchronous primary cancers of the endometrium and ovary: Do they have Lynch syndrome? J. Clin. Oncol. 2005, 23, 9344–9350. [Google Scholar] [CrossRef]
  35. Cederquist, K.; Emanuelsson, M.; Wiklund, F.; Golovleva, I.; Palmqvist, R.; Grönberg, H. Two Swedish founder MSH6 mutations, one nonsense and one missense, conferring high cumulative risk of Lynch syndrome. Clin. Genet. 2005, 68, 533–541. [Google Scholar] [CrossRef] [PubMed]
  36. Ollikainen, M.; Abdel-Rahman, W.M.; Moisio, A.-L.; Lindroos, A.; Kariola, R.; Järvelä, I.; Pöyhönen, M.; Butzow, R.; Peltomäki, P. Molecular analysis of familial endometrial carcinoma: A manifestation of hereditary nonpolyposis colorectal cancer or a separate syndrome? J. Clin. Oncol. 2005, 23, 4609–4616. [Google Scholar] [CrossRef]
  37. Taylor, N.P.; Zighelboim, I.; Huettner, P.C.; Powell, M.A.; Gibb, R.K.; Rader, J.S.; Mutch, D.G.; Edmonston, T.B.; Goodfellow, P.J. DNA mismatch repair and TP53 defects are early events in uterine carcinosarcoma tumorigenesis. Mod. Pathol. 2006, 19, 1333–1338. [Google Scholar] [CrossRef]
  38. Niessen, R.C.; Sijmons, R.H.; Ou, J.; Olthof, S.G.M.; Osinga, J.; Ligtenberg, M.J.; Hogervorst, F.B.L.; Weiss, M.M.; Tops, C.M.J.; Hes, F.J.; et al. MUTYH and the mismatch repair system: Partners in crime? Hum. Genet. 2006, 119, 206–211. [Google Scholar] [CrossRef] [Green Version]
  39. Rijcken, F.; van der Zee, A.; van der Sluis, T.; Boersma-van Ek, W.; Kleibeuker, J.; Hollema, H. Cell cycle regulators and apoptosis-associated proteins in relation to proliferative activity and degree of apoptosis in HNPCC versus sporadic endometrial carcinoma. Histopathology 2006, 48, 275–285. [Google Scholar] [CrossRef]
  40. Yoon, S.N.; Ku, J.-L.; Shin, Y.-K.; Kim, K.-H.; Choi, J.-S.; Jang, E.-J.; Park, H.-C.; Kim, D.-W.; Kim, M.A.; Kim, W.H.; et al. Hereditary nonpolyposis colorectal cancer in endometrial cancer patients. Int J. Cancer 2008, 122, 1077–1081. [Google Scholar] [CrossRef]
  41. Arabi, H.; Guan, H.; Kumar, S.; Cote, M.; Bandyopadhyay, S.; Bryant, C.; Shah, J.; Abdul-Karim, F.W.; Munkarah, A.R.; Ali-Fehmi, R. Impact of microsatellite instability (MSI) on survival in high grade endometrial carcinoma. Gynecol. Oncol. 2009, 113, 153–158. [Google Scholar] [CrossRef]
  42. Walsh, C.S.; Blum, A.; Walts, A.; Alsabeh, R.; Tran, H.; Koeffler, H.P.; Karlan, B.Y. Lynch syndrome among gynecologic oncology patients meeting Bethesda guidelines for screening. Gynecol. Oncol. 2010, 116, 516–521. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Yasue, A.; Hasegawa, K.; Udagawa, Y. Effects of tamoxifen on the endometrium and its mechanism of carcinogenicity. Hum. Cell 2011, 24, 65–73. [Google Scholar] [CrossRef] [PubMed]
  44. Huang, Y.-W.; Kuo, C.-T.; Chen, J.-H.; Goodfellow, P.J.; Huang, T.H.-M.; Rader, J.S.; Uyar, D.S. Hypermethylation of miR-203 in endometrial carcinomas. Gynecol. Oncol. 2014, 133, 340–345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Kobayashi, Y.; Nakamura, K.; Nomura, H.; Banno, K.; Irie, H.; Adachi, M.; Iida, M.; Umene, K.; Nogami, Y.; Masuda, K.; et al. Clinicopathologic analysis with immunohistochemistry for DNA mismatch repair protein expression in synchronous primary endometrial and ovarian cancers. Int. J. Gynecol. Cancer 2015, 25, 440–446. [Google Scholar] [CrossRef] [PubMed]
  46. Ren, C.; Liu, Y.; Wang, Y.; Tang, Y.; Wei, Y.; Liu, C.; Zhang, H. Identification of novel Lynch syndrome mutations in Chinese patients with endometriod endometrial cancer. Cancer Biol. Med. 2020, 17, 458–467. [Google Scholar] [CrossRef] [PubMed]
  47. Westin, S.N.; Lacour, R.A.; Urbauer, D.L.; Luthra, R.; Bodurka, D.C.; Lu, K.H.; Broaddus, R.R. Carcinoma of the lower uterine segment: A newly described association with Lynch syndrome. J. Clin. Oncol. 2008, 26, 5965–5971. [Google Scholar] [CrossRef]
  48. Matthews, K.S.; Estes, J.M.; Conner, M.G.; Manne, U.; Whitworth, J.M.; Huh, W.K.; Alvarez, R.D.; Straughn, J.M.; Barnes, M.N.; Rocconi, R.P. Lynch syndrome in women less than 50 years of age with endometrial cancer. Obstet. Gynecol. 2008, 111, 1161–1166. [Google Scholar] [CrossRef] [Green Version]
  49. Garg, K.; Shih, K.; Barakat, R.; Zhou, Q.; Iasonos, A.; Soslow, R.A. Endometrial carcinomas in women aged 40 years and younger: Tumors associated with loss of DNA mismatch repair proteins comprise a distinct clinicopathologic subset. Am. J. Surg. Pathol. 2009, 33, 1869–1877. [Google Scholar] [CrossRef] [PubMed]
  50. Garg, K.; Leitao, M.M.; Kauff, N.D.; Hansen, J.; Kosarin, K.; Shia, J.; Soslow, R.A. Selection of endometrial carcinomas for DNA mismatch repair protein immunohistochemistry using patient age and tumor morphology enhances detection of mismatch repair abnormalities. Am. J. Surg Pathol. 2009, 33, 925–933. [Google Scholar] [CrossRef]
  51. Tafe, L.J.; Garg, K.; Chew, I.; Tornos, C.; Soslow, R.A. Endometrial and ovarian carcinomas with undifferentiated components: Clinically aggressive and frequently underrecognized neoplasms. Mod. Pathol. 2010, 23, 781–789. [Google Scholar] [CrossRef]
  52. Cossio, S.L.; Koehler-Santos, P.; Pessini, S.A.; Mónego, H.; Edelweiss, M.I.; Meurer, L.; Errami, A.; Coffa, J.; Bock, H.; Saraiva-Pereira, M.L.; et al. Clinical and histomolecular endometrial tumor characterization of patients at-risk for Lynch syndrome in South of Brazil. Fam. Cancer 2010, 9, 131–139. [Google Scholar] [CrossRef]
  53. Resnick, K.E.; Frankel, W.L.; Morrison, C.D.; Fowler, J.M.; Copeland, L.J.; Stephens, J.; Kim, K.H.; Cohn, D.E. Mismatch repair status and outcomes after adjuvant therapy in patients with surgically staged endometrial cancer. Gynecol. Oncol. 2010, 117, 234–238. [Google Scholar] [CrossRef] [Green Version]
  54. Shih, K.K.; Garg, K.; Levine, D.A.; Kauff, N.D.; Abu-Rustum, N.R.; Soslow, R.A.; Barakat, R.R. Clinicopathologic significance of DNA mismatch repair protein defects and endometrial cancer in women 40years of age and younger. Gynecol. Oncol. 2011, 123, 88–94. [Google Scholar] [CrossRef]
  55. Leenen, C.H.M.; van Lier, M.G.F.; van Doorn, H.C.; van Leerdam, M.E.; Kooi, S.G.; de Waard, J.; Hoedemaeker, R.F.; van den Ouweland, A.M.W.; Hulspas, S.M.; Dubbink, H.J.; et al. Prospective evaluation of molecular screening for Lynch syndrome in patients with endometrial cancer ≤ 70 years. Gynecol. Oncol. 2012, 125, 414–420. [Google Scholar] [CrossRef]
  56. Soslow, R.A.; Wethington, S.L.; Cesari, M.; Chiappetta, D.; Olvera, N.; Shia, J.; Levine, D.A. Clinicopathologic analysis of matched primary and recurrent endometrial carcinoma. Am. J. Surg. Pathol. 2012, 36, 1771–1781. [Google Scholar] [CrossRef]
  57. Egoavil, C.; Alenda, C.; Castillejo, A.; Paya, A.; Peiro, G.; Sánchez-Heras, A.-B.; Castillejo, M.-I.; Rojas, E.; Barberá, V.-M.; Cigüenza, S.; et al. Prevalence of Lynch syndrome among patients with newly diagnosed endometrial cancers. PLoS ONE 2013, 8, e79737. [Google Scholar] [CrossRef] [Green Version]
  58. Bosse, T.; ter Haar, N.T.; Seeber, L.M.; v Diest, P.J.; Hes, F.J.; Vasen, H.F.A.; Nout, R.A.; Creutzberg, C.L.; Morreau, H.; Smit, V.T.H.B.M. Loss of ARID1A expression and its relationship with PI3K-Akt pathway alterations, TP53 and microsatellite instability in endometrial cancer. Mod. Pathol 2013, 26, 1525–1535. [Google Scholar] [CrossRef] [Green Version]
  59. Moline, J.; Mahdi, H.; Yang, B.; Biscotti, C.; Roma, A.A.; Heald, B.; Rose, P.G.; Michener, C.; Eng, C. Implementation of tumor testing for lynch syndrome in endometrial cancers at a large academic medical center. Gynecol. Oncol. 2013, 130, 121–126. [Google Scholar] [CrossRef]
  60. Peiró, G.; Peiró, F.M.; Ortiz-Martínez, F.; Planelles, M.; Sánchez-Tejada, L.; Alenda, C.; Ceballos, S.; Sánchez-Payá, J.; Laforga, J.B. Association of mammalian target of rapamycin with aggressive type II endometrial carcinomas and poor outcome: A potential target treatment. Hum. Pathol. 2013, 44, 218–225. [Google Scholar] [CrossRef]
  61. Romero-Pérez, L.; López-García, M.Á.; Díaz-Martín, J.; Biscuola, M.; Castilla, M.Á.; Tafe, L.J.; Garg, K.; Oliva, E.; Matias-Guiu, X.; Soslow, R.A.; et al. ZEB1 overexpression associated with E-cadherin and microRNA-200 downregulation is characteristic of undifferentiated endometrial carcinoma. Mod. Pathol. 2013, 26, 1514–1524. [Google Scholar] [CrossRef] [Green Version]
  62. Mills, A.M.; Liou, S.; Ford, J.M.; Berek, J.S.; Pai, R.K.; Longacre, T.A. Lynch syndrome screening should be considered for all patients with newly diagnosed endometrial cancer. Am. J. Surg. Pathol. 2014, 38, 1501–1509. [Google Scholar] [CrossRef] [PubMed]
  63. Ruiz, I.; Martín-Arruti, M.; Lopez-Lopez, E.; Garcia-Orad, A. Lack of association between deficient mismatch repair expression and outcome in endometrial carcinomas of the endometrioid type. Gynecol. Oncol. 2014, 134, 20–23. [Google Scholar] [CrossRef] [PubMed]
  64. Thoury, A.; Descatoire, V.; Kotelevets, L.; Kannengiesser, C.; Bertrand, G.; Theou-Anton, N.; Frey, C.; Genestie, C.; Raymond, E.; Chastre, E.; et al. Evidence for different expression profiles for c-Met, EGFR, PTEN and the mTOR pathway in low and high grade endometrial carcinomas in a cohort of consecutive women. Occurrence of PIK3CA and K-Ras mutations and microsatellite instability. Histol. Histopathol. 2014, 29, 1455–1466. [Google Scholar] [CrossRef] [PubMed]
  65. Rabban, J.T.; Calkins, S.M.; Karnezis, A.N.; Grenert, J.P.; Blanco, A.; Crawford, B.; Chen, L.-M. Association of tumor morphology with mismatch-repair protein status in older endometrial cancer patients: Implications for universal versus selective screening strategies for Lynch syndrome. Am. J. Surg. Pathol. 2014, 38, 793–800. [Google Scholar] [CrossRef]
  66. Long, Q.; Peng, Y.; Tang, Z.; Wu, C. Role of endometrial cancer abnormal MMR protein in screening Lynch-syndrome families. Int. J. Clin. Exp. Pathol. 2014, 7, 7297–7303. [Google Scholar]
  67. Woo, Y.L.; Cheah, P.L.; Shahruddin, S.I.; Omar, S.Z.; Arends, M. The immunohistochemistry signature of mismatch repair (MMR) proteins in a multiethnic Asian cohort with endometrial carcinoma. Int. J. Gynecol. Pathol. 2014, 33, 554–559. [Google Scholar] [CrossRef] [PubMed]
  68. Hoang, L.N.; Ali, R.H.; Lau, S.; Gilks, C.B.; Lee, C.-H. Immunohistochemical survey of mismatch repair protein expression in uterine sarcomas and carcinosarcomas. Int. J. Gynecol. Pathol. 2014, 33, 483–491. [Google Scholar] [CrossRef]
  69. Buchanan, D.D.; Tan, Y.Y.; Walsh, M.D.; Clendenning, M.; Metcalf, A.M.; Ferguson, K.; Arnold, S.T.; Thompson, B.A.; Lose, F.A.; Parsons, M.T.; et al. Tumor mismatch repair immunohistochemistry and DNA MLH1 methylation testing of patients with endometrial cancer diagnosed at age younger than 60 years optimizes triage for population-level germline mismatch repair gene mutation testing. J. Clin. Oncol. 2014, 32, 90–100. [Google Scholar] [CrossRef] [Green Version]
  70. Allo, G.; Bernardini, M.Q.; Wu, R.-C.; Shih, I.-M.; Kalloger, S.; Pollett, A.; Gilks, C.B.; Clarke, B.A. ARID1A loss correlates with mismatch repair deficiency and intact p53 expression in high-grade endometrial carcinomas. Mod. Pathol. 2014, 27, 255–261. [Google Scholar] [CrossRef] [Green Version]
  71. Goodfellow, P.J.; Billingsley, C.C.; Lankes, H.A.; Ali, S.; Cohn, D.E.; Broaddus, R.J.; Ramirez, N.; Pritchard, C.C.; Hampel, H.; Chassen, A.S.; et al. Combined Microsatellite Instability, MLH1 Methylation Analysis, and Immunohistochemistry for Lynch Syndrome Screening in Endometrial Cancers From GOG210: An NRG Oncology and Gynecologic Oncology Group Study. J. Clin. Oncol. 2015, 33, 4301–4308. [Google Scholar] [CrossRef]
  72. Chu, M.M.-Y.; Liu, S.S.; Tam, K.-F.; Ip, P.P.-C.; Cheung, A.N.-Y.; Ngan, H.Y.-S. The Significance of Mismatch Repair Deficiency in Young Patients With Endometrial Cancer. Int. J. Gynecol. Pathol. 2015, 34, 403–410. [Google Scholar] [CrossRef] [PubMed]
  73. Graham, R.P.; Kerr, S.E.; Butz, M.L.; Thibodeau, S.N.; Halling, K.C.; Smyrk, T.C.; Dina, M.A.; Waugh, V.M.; Rumilla, K.M. Heterogenous MSH6 loss is a result of microsatellite instability within MSH6 and occurs in sporadic and hereditary colorectal and endometrial carcinomas. Am. J. Surg. Pathol. 2015, 39, 1370–1376. [Google Scholar] [CrossRef] [PubMed]
  74. Dudley, B.; Brand, R.E.; Thull, D.; Bahary, N.; Nikiforova, M.N.; Pai, R.K. Germline MLH1 Mutations Are Frequently Identified in Lynch Syndrome Patients With Colorectal and Endometrial Carcinoma Demonstrating Isolated Loss of PMS2 Immunohistochemical Expression. Am. J. Surg. Pathol. 2015, 39, 1114–1120. [Google Scholar] [CrossRef]
  75. Stelloo, E.; Bosse, T.; Nout, R.A.; MacKay, H.J.; Church, D.N.; Nijman, H.W.; Leary, A.; Edmondson, R.J.; Powell, M.E.; Crosbie, E.J.; et al. Refining prognosis and identifying targetable pathways for high-risk endometrial cancer, a TransPORTEC initiative. Mod. Pathol. 2015, 28, 836–844. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Mao, T.-L.; Ayhan, A.; Kuo, K.-T.; Lin, M.-C.; Tseng, L.-H.; Ogawa, H. Immunohistochemical study of endometrial high-grade endometrioid carcinoma with or without a concurrent low-grade component: Implications for pathogenetic and survival differences. Histopathology 2015, 67, 474–482. [Google Scholar] [CrossRef] [PubMed]
  77. McConechy, M.K.; Talhouk, A.; Li-Chang, H.H.; Leung, S.; Huntsman, D.G.; Gilks, C.B.; McAlpine, J.N. Detection of DNA mismatch repair (MMR) deficiencies by immunohistochemistry can effectively diagnose the microsatellite instability (MSI) phenotype in endometrial carcinomas. Gynecol. Oncol. 2015, 137, 306–310. [Google Scholar] [CrossRef] [PubMed]
  78. Stewart, C.J.R.; Crook, M.L. SWI/SNF complex deficiency and mismatch repair protein expression in undifferentiated and dedifferentiated endometrial carcinoma. Pathology 2015, 47, 439–445. [Google Scholar] [CrossRef]
  79. Watkins, J.C.; Nucci, M.R.; Ritterhouse, L.L.; Howitt, B.E.; Sholl, L.M. Unusual Mismatch Repair Immunohistochemical Patterns in Endometrial Carcinoma. Am. J. Surg Pathol. 2016, 40, 909–916. [Google Scholar] [CrossRef]
  80. Pocrnich, C.E.; Ramalingam, P.; Euscher, E.D.; Malpica, A. Neuroendocrine Carcinoma of the Endometrium: A Clinicopathologic Study of 25 Cases. Am. J. Surg Pathol. 2016, 40, 577–586. [Google Scholar] [CrossRef]
  81. Lin, D.I.; Hecht, J.L. Targeted Screening With Combined Age- and Morphology-Based Criteria Enriches Detection of Lynch Syndrome in Endometrial Cancer. Int. J. Surg. Pathol. 2016, 24, 297–305. [Google Scholar] [CrossRef]
  82. Mills, A.M.; Sloan, E.A.; Thomas, M.; Modesitt, S.C.; Stoler, M.H.; Atkins, K.A.; Moskaluk, C.A. Clinicopathologic Comparison of Lynch Syndrome-associated and ‘Lynch-like’ Endometrial Carcinomas Identified on Universal Screening Using Mismatch Repair Protein Immunohistochemistry. Am. J. Surg. Pathol. 2016, 40, 155–165. [Google Scholar] [CrossRef] [PubMed]
  83. Ramalingam, P.; Masand, R.P.; Euscher, E.D.; Malpica, A. Undifferentiated Carcinoma of the Endometrium: An Expanded Immunohistochemical Analysis Including PAX-8 and Basal-Like Carcinoma Surrogate Markers. Int. J. Gynecol. Pathol. 2016, 35, 410–418. [Google Scholar] [CrossRef] [PubMed]
  84. Shikama, A.; Minaguchi, T.; Matsumoto, K.; Akiyama-Abe, A.; Nakamura, Y.; Michikami, H.; Nakao, S.; Sakurai, M.; Ochi, H.; Onuki, M.; et al. Clinicopathologic implications of DNA mismatch repair status in endometrial carcinomas. Gynecol. Oncol. 2016, 140, 226–233. [Google Scholar] [CrossRef] [PubMed]
  85. Kato, A.; Sato, N.; Sugawara, T.; Takahashi, K.; Kito, M.; Makino, K.; Sato, T.; Shimizu, D.; Shirasawa, H.; Miura, H.; et al. Isolated Loss of PMS2 Immunohistochemical Expression is Frequently Caused by Heterogenous MLH1 Promoter Hypermethylation in Lynch Syndrome Screening for Endometrial Cancer Patients. Am. J. Surg. Pathol. 2016, 40, 770–776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Okoye, E.I.; Bruegl, A.S.; Fellman, B.; Luthra, R.; Broaddus, R.R. Defective DNA Mismatch Repair Influences Expression of Endometrial Carcinoma Biomarkers. Int. J. Gynecol. Pathol. 2016, 35, 8–15. [Google Scholar] [CrossRef]
  87. Russo, M.; Broach, J.; Sheldon, K.; Houser, K.R.; Liu, D.J.; Kesterson, J.; Phaeton, R.; Hossler, C.; Hempel, N.; Baker, M.; et al. Clonal evolution in paired endometrial intraepithelial neoplasia/atypical hyperplasia and endometrioid adenocarcinoma. Hum. Pathol. 2017, 67, 69–77. [Google Scholar] [CrossRef] [PubMed]
  88. Bregar, A.; Deshpande, A.; Grange, C.; Zi, T.; Stall, J.; Hirsch, H.; Reeves, J.; Sathyanarayanan, S.; Growdon, W.B.; Rueda, B.R. Characterization of immune regulatory molecules B7-H4 and PD-L1 in low and high grade endometrial tumors. Gynecol. Oncol. 2017, 145, 446–452. [Google Scholar] [CrossRef] [Green Version]
  89. Pelletier, M.P.; Trinh, V.Q.; Stephenson, P.; Mes-Masson, A.-M.; Samouelian, V.; Provencher, D.M.; Rahimi, K. Microcystic, elongated, and fragmented pattern invasion is mainly associated with isolated tumor cell pattern metastases in International Federation of Gynecology and Obstetrics grade I endometrioid endometrial cancer. Hum. Pathol. 2017, 62, 33–39. [Google Scholar] [CrossRef] [PubMed]
  90. Stelloo, E.; Jansen, A.M.L.; Osse, E.M.; Nout, R.A.; Creutzberg, C.L.; Ruano, D.; Church, D.N.; Morreau, H.; Smit, V.T.H.B.M.; van Wezel, T.; et al. Practical guidance for mismatch repair-deficiency testing in endometrial cancer. Ann. Oncol. 2017, 28, 96–102. [Google Scholar] [CrossRef] [PubMed]
  91. Dillon, J.L.; Gonzalez, J.L.; DeMars, L.; Bloch, K.J.; Tafe, L.J. Universal screening for Lynch syndrome in endometrial cancers: Frequency of germline mutations and identification of patients with Lynch-like syndrome. Hum. Pathol. 2017, 70, 121–128. [Google Scholar] [CrossRef]
  92. Tangjitgamol, S.; Kittisiam, T.; Tanvanich, S. Prevalence and prognostic role of mismatch repair gene defect in endometrial cancer patients. Tumour. Biol. 2017, 39, 1010428317725834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Zakhour, M.; Cohen, J.G.; Gibson, A.; Walts, A.E.; Karimian, B.; Baltayan, A.; Aoyama, C.; Garcia, L.; Dhaliwal, S.K.; Elashoff, D.; et al. Abnormal mismatch repair and other clinicopathologic predictors of poor response to progestin treatment in young women with endometrial complex atypical hyperplasia and well-differentiated endometrial adenocarcinoma: A consecutive case series. BJOG 2017, 124, 1576–1583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Najdawi, F.; Crook, A.; Maidens, J.; McEvoy, C.; Fellowes, A.; Pickett, J.; Ho, M.; Nevell, D.; McIlroy, K.; Sheen, A.; et al. Lessons learnt from implementation of a Lynch syndrome screening program for patients with gynaecological malignancy. Pathology 2017, 49, 457–464. [Google Scholar] [CrossRef] [PubMed]
  95. Sloan, E.A.; Ring, K.L.; Willis, B.C.; Modesitt, S.C.; Mills, A.M. PD-L1 Expression in Mismatch Repair-deficient Endometrial Carcinomas, Including Lynch Syndrome-associated and MLH1 Promoter Hypermethylated Tumors. Am. J. Surg. Pathol. 2017, 41, 326–333. [Google Scholar] [CrossRef] [PubMed]
  96. Watkins, J.C.; Yang, E.J.; Muto, M.G.; Feltmate, C.M.; Berkowitz, R.S.; Horowitz, N.S.; Syngal, S.; Yurgelun, M.B.; Chittenden, A.; Hornick, J.L.; et al. Universal Screening for Mismatch-Repair Deficiency in Endometrial Cancers to Identify Patients With Lynch Syndrome and Lynch-like Syndrome. Int. J. Gynecol. Pathol. 2017, 36, 115–127. [Google Scholar] [CrossRef]
  97. Chen, W.; Husain, A.; Nelson, G.S.; Rambau, P.F.; Liu, S.; Lee, C.-H.; Lee, S.; Duggan, M.A.; Köbel, M. Immunohistochemical Profiling of Endometrial Serous Carcinoma. Int. J. Gynecol. Pathol. 2017, 36, 128–139. [Google Scholar] [CrossRef]
  98. Köbel, M.; Meng, B.; Hoang, L.N.; Almadani, N.; Li, X.; Soslow, R.A.; Gilks, C.B.; Lee, C.-H. Molecular Analysis of Mixed Endometrial Carcinomas Shows Clonality in Most Cases. Am. J. Surg. Pathol. 2016, 40, 166–180. [Google Scholar] [CrossRef]
  99. Pina, A.; Wolber, R.; McAlpine, J.N.; Gilks, B.; Kwon, J.S. Endometrial Cancer Presentation and Outcomes Based on Mismatch Repair Protein Expression From a Population-Based Study. Int. J. Gynecol. Cancer 2018, 28, 1624–1630. [Google Scholar] [CrossRef]
  100. Adar, T.; Rodgers, L.H.; Shannon, K.M.; Yoshida, M.; Ma, T.; Mattia, A.; Lauwers, G.Y.; Iafrate, A.J.; Hartford, N.M.; Oliva, E.; et al. Universal screening of both endometrial and colon cancers increases the detection of Lynch syndrome. Cancer 2018, 124, 3145–3153. [Google Scholar] [CrossRef] [Green Version]
  101. Chapel, D.B.; Yamada, S.D.; Cowan, M.; Lastra, R.R. Immunohistochemistry for mismatch repair protein deficiency in endometrioid endometrial carcinoma yields equivalent results when performed on endometrial biopsy/curettage or hysterectomy specimens. Gynecol. Oncol. 2018, 149, 570–574. [Google Scholar] [CrossRef]
  102. Bosse, T.; Nout, R.A.; McAlpine, J.N.; McConechy, M.K.; Britton, H.; Hussein, Y.R.; Gonzalez, C.; Ganesan, R.; Steele, J.C.; Harrison, B.T.; et al. Molecular Classification of Grade 3 Endometrioid Endometrial Cancers Identifies Distinct Prognostic Subgroups. Am. J. Surg. Pathol. 2018, 42, 561–568. [Google Scholar] [CrossRef] [PubMed]
  103. Saita, C.; Yamaguchi, T.; Horiguchi, S.-I.; Yamada, R.; Takao, M.; Iijima, T.; Wakaume, R.; Aruga, T.; Tabata, T.; Koizumi, K. Tumor development in Japanese patients with Lynch syndrome. PLoS ONE 2018, 13, e0195572. [Google Scholar] [CrossRef] [PubMed]
  104. Espinosa, I.; De Leo, A.; D’Angelo, E.; Rosa-Rosa, J.M.; Corominas, M.; Gonzalez, A.; Palacios, J.; Prat, J. Dedifferentiated endometrial carcinomas with neuroendocrine features: A clinicopathologic, immunohistochemical, and molecular genetic study. Hum. Pathol. 2018, 72, 100–106. [Google Scholar] [CrossRef] [PubMed]
  105. Li, Z.; Joehlin-Price, A.S.; Rhoades, J.; Ayoola-Adeola, M.; Miller, K.; Parwani, A.V.; Backes, F.J.; Felix, A.S.; Suarez, A.A. Programmed Death Ligand 1 Expression Among 700 Consecutive Endometrial Cancers: Strong Association With Mismatch Repair Protein Deficiency. Int. J. Gynecol. Cancer 2018, 28, 59–68. [Google Scholar] [CrossRef] [PubMed]
  106. Doghri, R.; Houcine, Y.; Boujelbène, N.; Driss, M.; Charfi, L.; Abbes, I.; Mrad, K.; Sellami, R. Mismatch Repair Deficiency in Endometrial Cancer, Immunohistochemistry Staining and Clinical Implications. Appl. Immunohistochem. Mol. Morphol. 2019, 27, 678–682. [Google Scholar] [CrossRef]
  107. Hashmi, A.A.; Mudassir, G.; Hashmi, R.N.; Irfan, M.; Asif, H.; Khan, E.Y.; Abu Bakar, S.M.; Faridi, N. Microsatellite Instability in Endometrial Carcinoma by Immunohistochemistry.y, Association with Clinical and Histopathologic Parameters. Asian Pac. J. Cancer Prev. 2019, 20, 2601–2606. [Google Scholar] [CrossRef] [PubMed]
  108. Saeki, H.; Hlaing, M.T.; Horimoto, Y.; Kajino, K.; Ohtsuji, N.; Fujino, K.; Terao, Y.; Hino, O. Usefulness of immunohistochemistry for mismatch repair protein and microsatellite instability examination in adenocarcinoma and background endometrium of sporadic endometrial cancer cases. J. Obstet. Gynaecol. Res. 2019, 45, 2037–2042. [Google Scholar] [CrossRef] [PubMed]
  109. Zannoni, G.F.; Santoro, A.; Angelico, G.; Spadola, S.; Arciuolo, D.; Valente, M.; Inzani, F.; Pettinato, A.; Vatrano, S.; Fanfani, F.; et al. Clear cell carcinoma of the endometrium: An immunohistochemical and molecular analysis of 45 cases. Hum. Pathol. 2019, 92, 10–17. [Google Scholar] [CrossRef]
  110. Abdulfatah, E.; Wakeling, E.; Sakr, S.; Al-Obaidy, K.; Bandyopadhyay, S.; Morris, R.; Feldman, G.; Ali-Fehmi, R. Molecular classification of endometrial carcinoma applied to endometrial biopsy specimens: Towards early personalized patient management. Gynecol. Oncol. 2019, 154, 467–474. [Google Scholar] [CrossRef]
  111. Chapel, D.B.; Patil, S.A.; Plagov, A.; Puranik, R.; Mendybaeva, A.; Steinhardt, G.; Wanjari, P.; Lastra, R.R.; Kadri, S.; Segal, J.P.; et al. Quantitative next-generation sequencing-based analysis indicates progressive accumulation of microsatellite instability between atypical hyperplasia/endometrial intraepithelial neoplasia and paired endometrioid endometrial carcinoma. Mod. Pathol. 2019, 32, 1508–1520. [Google Scholar] [CrossRef]
  112. Kahn, R.M.; Gordhandas, S.; Maddy, B.P.; Baltich Nelson, B.; Askin, G.; Christos, P.J.; Caputo, T.A.; Chapman-Davis, E.; Holcomb, K.; Frey, M.K. Universal endometrial cancer tumor typing: How much has immunohistochemistry, microsatellite instability, and MLH1 methylation improved the diagnosis of Lynch syndrome across the population? Cancer 2019, 125, 3172–3183. [Google Scholar] [CrossRef] [PubMed]
  113. Ryan, N.A.J.; Glaire, M.A.; Blake, D.; Cabrera-Dandy, M.; Evans, D.G.; Crosbie, E.J. The proportion of endometrial cancers associated with Lynch syndrome: A systematic review of the literature and meta-analysis. Genet. Med. 2019, 21, 2167–2180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Wu, X.; Snir, O.; Rottmann, D.; Wong, S.; Buza, N.; Hui, P. Minimal microsatellite shift in microsatellite instability high endometrial cancer: A significant pitfall in diagnostic interpretation. Mod. Pathol. 2019, 32, 650–658. [Google Scholar] [CrossRef] [PubMed]
  115. Saijo, M.; Nakamura, K.; Ida, N.; Nasu, A.; Yoshino, T.; Masuyama, H.; Yanai, H. Histologic Appearance and Immunohistochemistry of DNA Mismatch Repair Protein and p53 in Endometrial Carcinosarcoma: Impact on Prognosis and Insights Into Tumorigenesis. Am. J. Surg. Pathol. 2019, 43, 1493–1500. [Google Scholar] [CrossRef]
  116. Sarode, V.R.; Robinson, L. Screening for Lynch Syndrome by Immunohistochemistry of Mismatch Repair Proteins: Significance of Indeterminate Result and Correlation With Mutational Studies. Arch. Pathol. Lab. Med. 2019, 143, 1225–1233. [Google Scholar] [CrossRef] [Green Version]
  117. Sari, A.; Pollett, A.; Eiriksson, L.R.; Lumsden-Johanson, B.; Van de Laar, E.; Kazerouni, H.; Salehi, A.; Sur, M.; Lytwyn, A.; Ferguson, S.E. Interobserver Agreement for Mismatch Repair Protein Immunohistochemistry in Endometrial and Nonserous, Nonmucinous Ovarian Carcinomas. Am. J. Surg. Pathol. 2019, 43, 591–600. [Google Scholar] [CrossRef]
  118. Lucas, E.; Chen, H.; Molberg, K.; Castrillon, D.H.; Rivera Colon, G.; Li, L.; Hinson, S.; Thibodeaux, J.; Lea, J.; Miller, D.S.; et al. Mismatch Repair Protein Expression in Endometrioid Intraepithelial Neoplasia/Atypical Hyperplasia: Should We Screen for Lynch Syndrome in Precancerous Lesions? Int. J. Gynecol. Pathol. 2019, 38, 533–542. [Google Scholar] [CrossRef]
  119. Baniak, N.; Fadare, O.; Köbel, M.; DeCoteau, J.; Parkash, V.; Hecht, J.L.; Hanley, K.Z.; Gwin, K.; Zheng, W.; Quick, C.M.; et al. Targeted Molecular and Immunohistochemical Analyses of Endometrial Clear Cell Carcinoma Show that POLE Mutations and DNA Mismatch Repair Protein Deficiencies Are Uncommon. Am. J. Surg. Pathol. 2019, 43, 531–537. [Google Scholar] [CrossRef]
  120. Backes, F.J.; Haag, J.; Cosgrove, C.M.; Suarez, A.; Cohn, D.E.; Goodfellow, P.J. Mismatch repair deficiency identifies patients with high-intermediate-risk (HIR) endometrioid endometrial cancer at the highest risk of recurrence: A prognostic biomarker. Cancer 2019, 125, 398–405. [Google Scholar] [CrossRef] [Green Version]
  121. Dong, F.; Costigan, D.C.; Howitt, B.E. Targeted next-generation sequencing in the detection of mismatch repair deficiency in endometrial cancers. Mod. Pathol. 2019, 32, 252–257. [Google Scholar] [CrossRef]
  122. Gan, Q.; Crumley, S.; Broaddus, R.R. Molecular Modifiers of Hormone Receptor Action: Decreased Androgen Receptor Expression in Mismatch Repair Deficient Endometrial Endometrioid Adenocarcinoma. Int. J. Gynecol. Pathol. 2019, 38, 44–51. [Google Scholar] [CrossRef] [PubMed]
  123. He, Y.; Tao, X.; Huang, F.; Jia, N.; Du, Y.; Yu, J.; Feng, W. Clinicopathologic features of endometrial cancer in Chinese patients younger than 50 years with a family history of cancer. Medicine (Baltimore) 2018, 97, e12968. [Google Scholar] [CrossRef]
  124. Ryan, N.A.J.; McMahon, R.; Tobi, S.; Snowsill, T.; Esquibel, S.; Wallace, A.J.; Bunstone, S.; Bowers, N.; Mosneag, I.E.; Kitson, S.J.; et al. The proportion of endometrial tumours associated with Lynch syndrome (PETALS): A prospective cross-sectional study. PLoS Med. 2020, 17, e1003263. [Google Scholar] [CrossRef]
  125. Rosa, R.C.A.; Santis, J.O.; Teixeira, L.A.; Molfetta, G.A.; Dos Santos, J.T.T.; Ribeiro, V.D.S.; Chahud, F.; Ribeiro-Silva, A.; Brunaldi, M.O.; Silva, W.A.; et al. Lynch syndrome identification in a Brazilian cohort of endometrial cancer screened by a universal approach. Gynecol. Oncol. 2020, 159, 229–238. [Google Scholar] [CrossRef] [PubMed]
  126. Beinse, G.; Rance, B.; Just, P.-A.; Izac, B.; Letourneur, F.; Saidu, N.E.B.; Chouzenoux, S.; Nicco, C.; Goldwasser, F.; Batteux, F.; et al. Identification of TP53 mutated group using a molecular and immunohistochemical classification of endometrial carcinoma to improve prognostic evaluation for adjuvant treatments. Int. J. Gynecol. Cancer 2020, 30, 640–647. [Google Scholar] [CrossRef] [PubMed]
  127. Timmerman, S.; Van Rompuy, A.S.; Van Gorp, T.; Vanden Bempt, I.; Brems, H.; Van Nieuwenhuysen, E.; Han, S.N.; Neven, P.; Victoor, J.; Laenen, A.; et al. Analysis of 108 patients with endometrial carcinoma using the PROMISE classification and additional genetic analyses for MMR-D. Gynecol. Oncol. 2020, 157, 245–251. [Google Scholar] [CrossRef]
  128. Missaoui, N.; Boukhari, N.; Limam, S.; Hmissa, S.; Mokni, M. Utility of the immunohistochemical analysis of DNA mismatch-repair proteins in endometrial hyperplasia. Acta Histochem. 2020, 122, 151505. [Google Scholar] [CrossRef]
  129. Dasgupta, S.; Ewing-Graham, P.C.; Groenendijk, F.H.; Stam, O.; Biermann, K.E.; Doukas, M.; Dubbink, H.J.; van Velthuysen, M.F.; Dinjens, W.N.M.; Van Bockstal, M.R. Granular dot-like staining with MLH1 immunohistochemistry is a clone-dependent artefact. Pathol. Res. Pract. 2020, 216, 152581. [Google Scholar] [CrossRef]
  130. Kolehmainen, A.M.; Pasanen, A.M.; Koivisto-Korander, R.L.; Bützow, R.C.; Loukovaara, M.J. Molecular characterization in the prediction of disease extent in endometrial carcinoma. Eur. J. Obstet. Gynecol. Reprod Biol. 2021, 256, 478–483. [Google Scholar] [CrossRef]
  131. León-Castillo, A.; de Boer, S.M.; Powell, M.E.; Mileshkin, L.R.; Mackay, H.J.; Leary, A.; Nijman, H.W.; Singh, N.; Pollock, P.M.; Bessette, P.; et al. Molecular Classification of the PORTEC-3 Trial for High-Risk Endometrial Cancer, Impact on Prognosis and Benefit From Adjuvant Therapy. J. Clin. Oncol. 2020, 38, 3388–3397. [Google Scholar] [CrossRef]
  132. Rekhi, B.; Menon, S.; Deodhar, K.K.; Ghosh, J.; Chopra, S.; Maheshwari, A. Clinicopathological features of 50 mismatch repair (MMR)-deficient endometrial carcinomas, tested by immunohistochemistry: A single institutional feasibility study, India. Ann. Diagn Pathol. 2020, 47, 151558. [Google Scholar] [CrossRef]
  133. Kim, S.R.; Cloutier, B.T.; Leung, S.; Cochrane, D.; Britton, H.; Pina, A.; Storness-Bliss, C.; Farnell, D.; Huang, L.; Shum, K.; et al. Molecular subtypes of clear cell carcinoma of the endometrium: Opportunities for prognostic and predictive stratification. Gynecol. Oncol. 2020, 158, 3–11. [Google Scholar] [CrossRef] [PubMed]
  134. Pasanen, A.; Ahvenainen, T.; Pellinen, T.; Vahteristo, P.; Loukovaara, M.; Bützow, R. PD-L1 Expression in Endometrial Carcinoma Cells and Intratumoral Immune Cells: Differences Across Histologic and TCGA-based Molecular Subgroups. Am. J. Surg. Pathol. 2020, 44, 174–181. [Google Scholar] [CrossRef] [PubMed]
  135. Jin, C.; Hacking, S.; Liang, S.; Nasim, M. PD-L1/PD-1 Expression in Endometrial Clear Cell Carcinoma: A Potential Surrogate Marker for Clinical Trials. Int. J. Surg. Pathol. 2020, 28, 31–37. [Google Scholar] [CrossRef]
  136. Rowe, M.; Krishnan, R.; Mills, A.; Ring, K. β-catenin and PD-L1 expression in mismatch repair deficient endometrial carcinomas. Int. J. Gynecol. Cancer 2020, 30, 993–999. [Google Scholar] [CrossRef] [PubMed]
  137. Stinton, C.; Fraser, H.; Al-Khudairy, L.; Court, R.; Jordan, M.; Grammatopoulos, D.; Taylor-Phillips, S. Testing for lynch syndrome in people with endometrial cancer using immunohistochemistry and microsatellite instability-based testing strategies-A systematic review of test accuracy. Gynecol. Oncol. 2021, 160, 148–160. [Google Scholar] [CrossRef]
  138. Pécriaux, A.; Favre, L.; Calderaro, J.; Charpy, C.; Derman, J.; Pujals, A. Detection of microsatellite instability in a panel of solid tumours with the Idylla MSI Test using extracted DNA. J. Clin. Pathol. 2021, 74, 36–42. [Google Scholar] [CrossRef]
  139. Tjalsma, A.S.; Wagner, A.; Dinjens, W.N.M.; Ewing-Graham, P.C.; Alcalá, L.S.M.; de Groot, M.E.R.; Hamoen, K.E.; van Hof, A.C.; Hofhuis, W.; Hofman, L.N.; et al. Evaluation of a nationwide Dutch guideline to detect Lynch syndrome in patients with endometrial cancer. Gynecol. Oncol. 2021, 160, 771–776. [Google Scholar] [CrossRef]
  140. Joehlin-Price, A.; Van Ziffle, J.; Hills, N.K.; Ladwig, N.; Rabban, J.T.; Garg, K. Molecularly Classified Uterine FIGO Grade 3 Endometrioid Carcinomas Show Distinctive Clinical Outcomes But Overlapping Morphologic Features. Am. J. Surg. Pathol. 2021, 45, 421–429. [Google Scholar] [CrossRef]
  141. Yamamoto, A.; Yamaguchi, T.; Suzuki, O.; Ito, T.; Chika, N.; Kamae, N.; Tamaru, J.-I.; Nagai, T.; Seki, H.; Arai, T.; et al. Prevalence and molecular characteristics of DNA mismatch repair deficient endometrial cancer in a Japanese hospital-based population. Jpn J. Clin. Oncol. 2021, 51, 60–69. [Google Scholar] [CrossRef]
  142. McConechy, M.K.; Talhouk, A.; Leung, S.; Chiu, D.; Yang, W.; Senz, J.; Reha-Krantz, L.J.; Lee, C.-H.; Huntsman, D.G.; Gilks, C.B.; et al. Endometrial Carcinomas with POLE Exonuclease Domain Mutations Have a Favorable Prognosis. Clin. Cancer Res. 2016, 22, 2865–2873. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Horowitz, N.; Pinto, K.; Mutch, D.G.; Herzog, T.J.; Rader, J.S.; Gibb, R.; Bocker-Edmonston, T.; Goodfellow, P.J. Microsatellite instability, MLH1 promoter methylation, and loss of mismatch repair in endometrial cancer and concomitant atypical hyperplasia. Gynecol. Oncol. 2002, 86, 62–68. [Google Scholar] [CrossRef] [PubMed]
  144. Strazzullo, M.; Cossu, A.; Baldinu, P.; Colombino, M.; Satta, M.P.; Tanda, F.; De Bonis, M.L.; Cerase, A.; D’Urso, M.; D’Esposito, M.; et al. High-resolution methylation analysis of the hMLH1 promoter in sporadic endometrial and colorectal carcinomas. Cancer 2003, 98, 1540–1546. [Google Scholar] [CrossRef]
  145. Kanaya, T.; Kyo, S.; Sakaguchi, J.; Maida, Y.; Nakamura, M.; Takakura, M.; Hashimoto, M.; Mizumoto, Y.; Inoue, M. Association of mismatch repair deficiency with PTEN frameshift mutations in endometrial cancers and the precursors in a Japanese population. Am. J. Clin. Pathol. 2005, 124, 89–96. [Google Scholar] [CrossRef] [PubMed]
  146. Zighelboim, I.; Powell, M.A.; Babb, S.A.; Whelan, A.J.; Schmidt, A.P.; Clendenning, M.; Senter, L.; Thibodeau, S.N.; de la Chapelle, A.; Goodfellow, P.J. Epitope-positive truncating MLH1 mutation and loss of PMS2: Implications for IHC-directed genetic testing for Lynch syndrome. Fam. Cancer 2009, 8, 501–504. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Koyamatsu, Y.; Sakamoto, M.; Miyake, K.; Muroya, T.; Sugano, K.; Nakao, Y.; Yokoyama, M.; Iwasaka, T. Gene expression profiles and microsatellite instability in uterine corpus endometrioid adenocarcinoma. J. Obstet. Gynaecol. Res. 2010, 36, 336–343. [Google Scholar] [CrossRef]
  148. Batte, B.A.L.; Bruegl, A.S.; Daniels, M.S.; Ring, K.L.; Dempsey, K.M.; Djordjevic, B.; Luthra, R.; Fellman, B.M.; Lu, K.H.; Broaddus, R.R. Consequences of universal MSI/IHC in screening ENDOMETRIAL cancer patients for Lynch syndrome. Gynecol. Oncol. 2014, 134, 319–325. [Google Scholar] [CrossRef] [Green Version]
  149. Bruegl, A.S.; Djordjevic, B.; Urbauer, D.L.; Westin, S.N.; Soliman, P.T.; Lu, K.H.; Luthra, R.; Broaddus, R.R. Utility of MLH1 methylation analysis in the clinical evaluation of Lynch Syndrome in women with endometrial cancer. Curr. Pharm. Des. 2014, 20, 1655–1663. [Google Scholar] [CrossRef] [Green Version]
  150. Goverde, A.; Spaander, M.C.; van Doorn, H.C.; Dubbink, H.J.; van den Ouweland, A.M.; Tops, C.M.; Kooi, S.G.; de Waard, J.; Hoedemaeker, R.F.; Bruno, M.J.; et al. Cost-effectiveness of routine screening for Lynch syndrome in endometrial cancer patients up to 70years of age. Gynecol. Oncol. 2016, 143, 453–459. [Google Scholar] [CrossRef]
  151. Bruegl, A.S.; Kernberg, A.; Broaddus, R.R. Importance of PCR-based Tumor Testing in the Evaluation of Lynch Syndrome-associated Endometrial Cancer. Adv. Anat. Pathol. 2017, 24, 372–378. [Google Scholar] [CrossRef]
  152. Zeimet, A.G.; Mori, H.; Petru, E.; Polterauer, S.; Reinthaller, A.; Schauer, C.; Scholl-Firon, T.; Singer, C.; Wimmer, K.; Zschocke, J.; et al. AGO Austria recommendation on screening and diagnosis of Lynch syndrome (LS). Arch. Gynecol. Obstet. 2017, 296, 123–127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Parc, Y.R.; Halling, K.C.; Burgart, L.J.; McDonnell, S.K.; Schaid, D.J.; Thibodeau, S.N.; Halling, A.C. Microsatellite instability and hMLH1/hMSH2 expression in young endometrial carcinoma patients: Associations with family history and histopathology. Int. J. Cancer 2000, 86, 60–66. [Google Scholar] [CrossRef]
  154. Ju, W.; Park, H.M.; Lee, S.N.; Sung, S.H.; Kim, S.C. Loss o.of hMLH1 expression is associated with less aggressive clinicopathological features in sporadic endometrioid endometrial adenocarcinoma. J. Obstet. Gynaecol. Res. 2006, 32, 454–460. [Google Scholar] [CrossRef] [PubMed]
  155. Choi, Y.D.; Choi, J.; Kim, J.H.; Lee, J.S.; Lee, J.H.; Choi, C.; Choi, H.S.; Lee, M.C.; Park, C.S.; Juhng, S.W.; et al. Microsatellite instability at a tetranucleotide repeat in type I endometrial carcinoma. J. Exp. Clin. Cancer Res. 2008, 27, 88. [Google Scholar] [CrossRef] [Green Version]
  156. Catasus, L.; D’Angelo, E.; Pons, C.; Espinosa, I.; Prat, J. Expression profiling of 22 genes involved in the PI3K-AKT pathway identifies two subgroups of high-grade endometrial carcinomas with different molecular alterations. Mod. Pathol. 2010, 23, 694–702. [Google Scholar] [CrossRef]
  157. An, H.J.; Kim, K.I.; Kim, J.Y.; Shim, J.Y.; Kang, H.; Kim, T.H.; Kim, J.K.; Jeong, J.K.; Lee, S.Y.; Kim, S.J. Microsatellite instability in endometrioid type endometrial adenocarcinoma is associated with poor prognostic indicators. Am. J. Surg Pathol. 2007, 31, 846–853. [Google Scholar] [CrossRef]
  158. Kolin, D.L.; Quick, C.M.; Dong, F.; Fletcher, C.D.M.; Stewart, C.J.R.; Soma, A.; Hornick, J.L.; Nucci, M.R.; Howitt, B.E. SMARCA4-deficient Uterine Sarcoma and Undifferentiated Endometrial Carcinoma Are Distinct Clinicopathologic Entities. Am. J. Surg. Pathol. 2020, 44, 263–270. [Google Scholar] [CrossRef]
  159. Schröer, A.; Köster, F.; Fischer, D.; Dubitscher, R.M.; Woll-Hermann, A.; Diedrich, K.; Friedrich, M.; Salehin, D. Immunohistochemistry of DNA mismatch repair enzyme MSH2 is not correlated with prognostic data from endometrial carcinomas. Anticancer Res. 2009, 29, 4833–4837. [Google Scholar]
  160. Salvesen, H.B.; MacDonald, N.; Ryan, A.; Jacobs, I.J.; Lynch, E.D.; Akslen, L.A.; Das, S. PTEN methylation is associated with advanced stage and microsatellite instability in endometrial carcinoma. Int. J. Cancer 2001, 91, 22–26. [Google Scholar] [CrossRef]
  161. Jones, N.L.; Xiu, J.; Rocconi, R.P.; Herzog, T.J.; Winer, I.S. Immune checkpoint expression, microsatellite instability, and mutational burden: Identifying immune biomarker phenotypes in uterine cancer. Gynecol. Oncol. 2020, 156, 393–399. [Google Scholar] [CrossRef]
  162. Colle, R.; Cohen, R.; Cochereau, D.; Duval, A.; Lascols, O.; Lopez-Trabada, D.; Afchain, P.; Trouilloud, I.; Parc, Y.; Lefevre, J.H.; et al. Immunotherapy and patients treated for cancer with microsatellite instability. Bull. Cancer 2017, 104, 42–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Leach, F.S.; Polyak, K.; Burrell, M.; Johnson, K.A.; Hill, D.; Dunlop, M.G.; Wyllie, A.H.; Peltomaki, P.; de la Chapelle, A.; Hamilton, S.R.; et al. Expression of the human mismatch repair gene hMSH2 in normal and neoplastic tissues. Cancer Res. 1996, 56, 235–240. [Google Scholar]
  164. Fishel, R.; Lescoe, M.K.; Rao, M.R.; Copeland, N.G.; Jenkins, N.A.; Garber, J.; Kane, M.; Kolodner, R. The human mutator gene homolog MSH2 and its association with hereditary nonpolyposis colon cancer. Cell 1993, 75, 1027–1038. [Google Scholar] [CrossRef]
  165. Bronner, C.E.; Baker, S.M.; Morrison, P.T.; Warren, G.; Smith, L.G.; Lescoe, M.K.; Kane, M.; Earabino, C.; Lipford, J.; Lindblom, A.; et al. Mutation in the DNA mismatch repair gene homologue hMLH 1 is associated with hereditary non-polyposis colon cancer. Nature 1994, 368, 258–261. [Google Scholar] [CrossRef] [PubMed]
  166. Nicolaides, N.C.; Papadopoulos, N.; Liu, B.; Weit, Y.-F.; Carter, K.C.; Ruben, S.M.; Rosen, C.A.; Haseltine, W.A.; Fleischmann, R.D.; Fraser, C.M.; et al. Mutations of two P/WS homologues in hereditary nonpolyposis colon cancer. Nature 1994, 371, 75–80. [Google Scholar] [CrossRef] [PubMed]
  167. Devlin, L.A.; Graham, C.A.; Price, J.H.; Morrison, P.J. Germline MSH6 mutations are more prevalent in endometrial cancer patient cohorts than hereditary non polyposis colorectal cancer cohorts. Ulster Med. J. 2008, 77, 25–30. [Google Scholar]
  168. Kawaguchi, M.; Banno, K.; Yanokura, M.; Kobayashi, Y.; Kishimi, A.; Ogawa, S.; Kisu, I.; Nomura, H.; Hirasawa, A.; Susumu, N.; et al. Analysis of candidate target genes for mononucleotide repeat mutation in microsatellite instability-high (MSI-H) endometrial cancer. Int. J. Oncol. 2009, 35, 977–982. [Google Scholar] [CrossRef] [Green Version]
  169. Lemetre, C.; Vieites, B.; Ng, C.K.Y.; Piscuoglio, S.; Schultheis, A.M.; Marchiò, C.; Murali, R.; Lopez-García, M.A.; Palacios, J.C.; Jungbluth, A.A.; et al. RNASeq analysis reveals biological processes governing the clinical behaviour of endometrioid and serous endometrial cancers. Eur. J. Cancer 2016, 64, 149–158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Armstrong, D.K.; Alvarez, R.D.; Bakkum-Gamez, J.N.; Barroilhet, L.; Behbakht, K.; Berchuck, A.; Berek, J.S.; Chen, L.-M.; Cristea, M.; DeRosa, M.; et al. NCCN Guidelines Insights: Ovarian Cancer, Version 1.2019. J. Natl. Compr. Canc. Netw. 2019, 17, 896–909. [Google Scholar] [CrossRef] [Green Version]
  171. Manchanda, R.; Menon, U.; Michaelson-Cohen, R.; Beller, U.; Jacobs, I. Hereditary non-polyposis colorectal cancer or Lynch syndrome: The gynaecological perspective. Curr. Opin. Obstet. Gynecol. 2009, 21, 31–38. [Google Scholar] [CrossRef]
  172. Senter, L.; Clendenning, M.; Sotamaa, K.; Hampel, H.; Green, J.; Potter, J.D.; Lindblom, A.; Lagerstedt, K.; Thibodeau, S.N.; Lindor, N.M.; et al. The clinical phenotype of Lynch syndrome due to germ-line PMS2 mutations. Gastroenterology 2008, 135, 419–428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Snowsill, T.M.; Ryan, N.A.J.; Crosbie, E.J.; Frayling, I.M.; Evans, D.G.; Hyde, C.J. Cost-effectiveness analysis of reflex testing for Lynch syndrome in women with endometrial cancer in the UK setting. PLoS ONE 2019, 14, e0221419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Gilson, P.; Levy, J.; Rouyer, M.; Demange, J.; Husson, M.; Bonnet, C.; Salleron, J.; Leroux, A.; Merlin, J.-L.; Harlé, A. Evaluation of 3 molecular-based assays for microsatellite instability detection in formalin-fixed tissues of patients with endometrial and colorectal cancers. Sci. Rep. 2020, 10, 16386. [Google Scholar] [CrossRef]
  175. Libera, L.; Craparotta, I.; Sahnane, N.; Chiaravalli, A.M.; Mannarino, L.; Cerutti, R.; Riva, C.; Marchini, S.; Furlan, D. Targeted gene sequencing of Lynch syndrome-related and sporadic endometrial carcinomas. Hum. Pathol. 2018, 81, 235–244. [Google Scholar] [CrossRef]
  176. Wolchok, J.D.; Chiarion-Sileni, V.; Gonzalez, R.; Rutkowski, P.; Grob, J.-J.; Cowey, C.L.; Lao, C.D.; Wagstaff, J.; Schadendorf, D.; Ferrucci, P.F.; et al. Overall Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2017, 377, 1345–1356. [Google Scholar] [CrossRef]
  177. Motzer, R.J.; Tannir, N.M.; McDermott, D.F.; Arén Frontera, O.; Melichar, B.; Choueiri, T.K.; Plimack, E.R.; Barthélémy, P.; Porta, C.; George, S.; et al. Nivolumab plus Ipilimumab versus Sunitinib in Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 2018, 378, 1277–1290. [Google Scholar] [CrossRef]
  178. Di Tucci, C.; Capone, C.; Galati, G.; Iacobelli, V.; Schiavi, M.C.; Di Donato, V.; Muzii, L.; Panici, P.B. Immunotherapy in endometrial cancer: New scenarios on the horizon. J. Gynecol. Oncol. 2019, 30, e46. [Google Scholar] [CrossRef] [Green Version]
  179. Nishio, H.; Iwata, T.; Aoki, D. Current status of cancer immunotherapy for gynecologic malignancies. Jpn. J. Clin. Oncol. 2021, 51, 167–172. [Google Scholar] [CrossRef]
  180. Taylor, M.H.; Lee, C.-H.; Makker, V.; Rasco, D.; Dutcus, C.E.; Wu, J.; Stepan, D.E.; Shumaker, R.C.; Motzer, R.J. Phase IB/II Trial of Lenvatinib Plus Pembrolizumab in Patients With Advanced R.Renal Cell Carcinoma, Endometrial Cancer, and Other Selected Advanced Solid Tumors. J. Clin. Oncol. 2020, 38, 1154–1163. [Google Scholar] [CrossRef]
  181. Marabelle, A.; Fakih, M.; Lopez, J.; Shah, M.; Shapira-Frommer, R.; Nakagawa, K.; Chung, H.C.; Kindler, H.L.; Lopez-Martin, J.A.; Miller, W.H.; et al. Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: Prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol. 2020, 21, 1353–1365. [Google Scholar] [CrossRef]
  182. Jumaah, A.S.; Al-Haddad, H.S.; Salem, M.M.; McAllister, K.A.; Yasseen, A.A. Mismatch repair deficiency and clinicopathological characteristics in endometrial carcinoma: A systematic review and meta-analysis. J. Pathol. Transl. Med. 2021, 55, 202–211. [Google Scholar] [CrossRef]
  183. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef]
Figure 1. PRISMA flowchart.
Figure 1. PRISMA flowchart.
Cancers 14 03783 g001
Table 1. Differences in immunohistochemistry protein losses in tumors with microinstability in endometrial cancer using the expression of two proteins.
Table 1. Differences in immunohistochemistry protein losses in tumors with microinstability in endometrial cancer using the expression of two proteins.
ReferenceType of TissueGerminal Mutation IncludedMMRd (n)MMRd (%)MLH1 (%)MSH2 (%)MSH6 (%)PMS2 (%)MLH1 Promoter Methylation (%)
Simpkins 1999 [19]FPPE + frozen tissue05310014.386--77
Peiro 2001 [20]FFPE01213.512.41.1---
Maruyama 2001 [21]FFPE131371219---
Berends 2001 [22]FFPE1132761.530.7---
Chiaravalli 2001 [23]FFPE11339.4-----
Hardisson 2003 [24]FFPE02329.573.921.7---
Sutter 2004 [25]Frozen tissue11339.42076.9---
Irving 2005 [26]FFPE065083.316.7--83
Alvarez 2012 [27]FFPE + frozen Tissue04167525---
Plotkin 2020 [28]FFPE01122--0100-
FFPE: formalin-fixed paraffin-embedded, MMRd: mismatch repair-deficient.
Table 2. Differences in immunohistochemistry protein losses in tumors with microinstability in endometrial cancer using the expression of three proteins.
Table 2. Differences in immunohistochemistry protein losses in tumors with microinstability in endometrial cancer using the expression of three proteins.
ReferenceType of Tissue Germinal Mutation IncludedMMRd (n)MMRd (%)MLH1 (%)MSH2 (%)MSH6 (%)PMS2 (%)MLH1 Promoter Methylation
Planck 2002 [29]FFPE121-9.547.657.1--
Orbo 2003 [30]FFPE018-55.522.233.3--
Lipton 2004 [31]Healthy tissue1672744.843.31.5--
Macdonald 2004 [32]FFPE + frozen016451.9141917-69
Buttin 2004 [33]Frozen tissue + healthy tissue19422.8----70
Soliman 2005 [34]FPPE1122058.341.741.7--
Cederquist 2005 [35]FFPE161000066.7--
Ollikainen 2005 [36]FFPE + healthy tissue11648.543.82531.3--
Taylor 2006 [37]FPPE062133.300--
Niessen 2006 [38]Not described03617.130.613.955.6--
Rijcken 2006 [39]FFPE11810033.311.111.1--
Yoon 2008 [40]FFPE + healthy tissue15044.24642-14
Arabi 2009 [41]FFPE + healthy tissue0252122.51843--
Walsh 2010 [42]FFPE1912.51226--
Yasue 2011 [43]FFPE + frozen tissue0822.91002545.8--
Huang 2014 [44]FFPE129-27.655.217.2--
Kobayashi 2015 [45]FFPE01753.182.411.864.7--
Ren 2020 [46]FFPE12712.838.13751.8-33.3
FFPE: formalin-fixed paraffin-embedded, MMRd: mismatch repair-deficient.
Table 3. Differences in immunohistochemistry protein losses in tumors with microinstability in endometrial cancer using expression of four proteins.
Table 3. Differences in immunohistochemistry protein losses in tumors with microinstability in endometrial cancer using expression of four proteins.
ReferenceType of Tissue Germinal Mutation IncludedMMRd (n)MMRd (%)MLH1 (%)MSH2 (%)MSH6 (%)PMS2 (%)MLH1 Promoter Methylation (%)
Westin 2008 [47]FFPE11234.32575752516.7
Matthews 2008 [48]FFPE + healthy tissue12134.485.795.323.876.2-
Garg 2009 [49]FFPE092033.333.35044.4-
Garg 2009 [50]FFPE1324559.440.640.659.4-
Tafe 2010 [51]Not described184787.5012.587.5-
Cossio 2010 [52]FFPE + healthy tissue173028.614.371.428.6-
Resnick 2010 [53]FFPE015566.5 ----
Shih 2011 [54]FFPE1916.144.455.655.644.4-
Leenen 2012 [55]FFPE14223.576.223.823.823.873.8
Soslow 2012 [56]FFPE0731.810000100-
Egoavil 2013 [57]FFPE16133.572.18.221.313.155.7
Bosse 2013 [58]FFPE13624.7----88.9
Moline 2013 [59]FFPE15924.184.715.318.615.355.9
Peiro 2013 [60]FFPE16324.479.44.817.679.4-
Romero-Perez 2013 [61]FFPE03932.548.723.130.869.2-
Mills 2014 [62]FFPE 113722.672.3272772.3-
Ruiz 2014 [63]FFPE06430.254.76.354.756.3-
Thoury 2014 [64]FFPE + healthy tissue01724.66502359-
Rabban 2014 [65]FFPE 1411575.67.7177.3-
Long 2014 [66]Not described14123.724.451.268.331.7-
Woo 2014 [67]FFPE01519.586.713.313.386.7-
Hoang 2014 [68]FFPE069.55016.716.783.3-
Buchanan 2014 [69]FFPE117024751324.775.3-
Allo 2014 [70]FFPE063337315.923.873-
Goodfellow 2015 [71]FFPE + Frozen tissue136038.475.23.111.97.570.3
Chu 2015 [72]FFPE + frozen tissue + healthy tissue02232.827.222.772.327.2-
Graham 2015 [73]FFPE + healthy tissue1-------
Dudley 2015 [74]FFPE17233.4---20.8-
Stelloo 2015 [75]FFPE01916.463.2---47.4
Mao 2015 [76]Not described01946.373.710.515.878.9-
Mc Conechy 2015 [77]Frozen tissue03824.2505.310.555.3-
Stewart 2015 [78]FFPE01359.176.915.430.884.7-
Watkins 2016 [79]FFPE12721.688.93.714.844.425.9
Pocrnich 2016 [80]Not described1844.47512.512.575-
Lin 2016 [81]Not described11722.382.317.617.682.364.7
Mills 2016 [82]Not described16631.465.218.231.871.2-
Ramalingam 2016 [83]FFPE01851.494.45.65.694.4-
Shikama 2016 [84]FFPE1622862.914.538.767.7-
Kato 2016 [85]FFPE182.2-----
Okoye 2016 [86]FFPE0409.7----75
Russo 2017 [87]FFPE + healthy tissue035066.733.333.366.7-
Bregar 2017 [88]FFPE01318.5-00--
Pelletier 2017 [89]Not described03426.867.68.814.785.3-
Stelloo 2017 [90]FFPE016924.385.25.913.689.9-
Dillon 2017 [91]FFPE16026853.33.38581.2
Tangjitgamol 2017 [92]FFPE021255.160.429.770.362.3-
Zakhour 2017 [93]FFPE167.133.35066.733.3-
Najdawi 2017 [94]FFPE1362986.713.926.793.3-
Sloan 2017 [95]FFPE13856.7----15.8
Watkins 2017 [96]FFPE14819.881.38.314.685.4-
Chen 2017 [97]FFPE03010.3-----
Kobel 2017 [98]FFPE0637.55033.35033.3-
Pina 2018 [99]Not described124227.178.921.121.178.969
Adar 2018 [100]Not described110722.180.319.619.615.470
Chapel 2018 [101]FFPE13030.386.7101090-
Bosse 2018 [102]Not described013636.2-----
Saita 2018 [103]Not described113-46.223.130.8--
Espinosa 2018 [104]FFPE 025010000100-
Li 2018 [105]FFPE016223.180.219.819.880.2-
Doghri 2019 [106]FFPE01022.280101080-
Hashmi 2019 [107]FFPE15644.492.917.935.789.3-
Saeki 2019 [108]FFPE + healthy tissue01818.477.822.244.483.3-
Zannoni 2019 [109]FFPE 01533.3046.773.326.7-
Abdufatah 2019 [110]Not described020405108080-
Chapel 2019 [111]FFPE11710094.15.95.994.1-
Kahn 2019 [112]Not described1167228.369.3---53.9
Ryan 2019 [113]Not described1256324.517.533237.6
Wu 2019 [114]FFPE15010063102472-
Saijo 2019 [115]FFPE 0610.5-----
Sarode 2019 [116]FFPE1459.3----26.7
Sari 2019 [117]FFPE0223068.2913.668.2-
Lucas 2019 [118]FFPE163-541942.955.6-
Baniak 2019 [119]FFPE 00------
Backes 2019 [120]Not described06432.5-----
Dong 2019 [121]FFPE 06324-----
Gan 2019 [122]FFPE19127.287.912.112.187.969.2
He 2019 [123]Not described123.3 -
Ryan 2020 [124]FFPE + healthy tissue11322675.89.118.975.862.9
Rosa 2020 [125]FFPE 18033.151.312.522.543.848.8
Beinse 2020 [126]FFPE03529.7--1783-
Timmerman 2020 [127]Not described1333181.8315.26.179
Missaoui 2020 [128]FFPE113.7100--100100
Dasgupta 2020 [129]Not described14-100--100-
Kolehmainen 2020 [130]FFPE028747.5-----
León-Castillo 2020 [131]FFPE013733.4-----
Rekhi 2020 [132]FFPE 050-66282866-
Kim 2020 [133]FFPE059.6-----
Pasanen 2020 [134]FFPE + tumor cells019137.3-----
Jin 2020 [135]Not described015-----
Rowe 2020 [136]FFPE143-----46.5
Stinton 2021 [137]Not described1-------
Pecriaux 2021 [138]FFPE19601000088.9-
Tjalsma 2021 [139]Not described1412314991414
Joehlin-Price 2021 [140]FFPE13536.877.111.425.977.120
Yamamoto 2021 [141]FFPE16817.277.916.217.679.475
FFPE: formalin-fixed paraffin-embedded, MMRd: mismatch repair-deficient.
Table 4. Differences in MLH1 promoter methylation.
Table 4. Differences in MLH1 promoter methylation.
ReferenceTechniqueMLH1 Promoter Methylation (%)
Simpkins 1999 [19]PCR + bisulfite conversion14.3
Horowitz 2002 [143]PCR + bisulfite conversion-
Strazzullo 2003 [144]PCR-
Buttin 2004 [33]PCR + bisulfite conversion70
Macdonald 2004 [32]Not described 69
Soliman 2005 [34]PCR + bisulfite conversion-
Irving 2005 [26]PCR83.3
Kanaya 2005 [145]PCR + bisulfite conversion-
Ollikainen 2005 [36]PCR-
Westin 2008 [47]PCR16.7
Nam Yoon 2008 [40]PCR14
Zighelboim 2009 [146]Pyrosequencing and/or combined bisulfite restriction analysis
Koyamatsu 2010 [147]Not described -
Walsh 2010 [42]PCR + bisulfite conversion-
Cossio 2010 [52]Methylation-specific multiplex ligation-dependent probe amplification
Leenen 2012 [55]Methylation-specific multiplex ligation-dependent probe amplification73.8
Egoavil 2013 [57]Methylation-specific multiplex ligation-dependent probe amplification55.7
Bosse 2013 [58]PCR + bisulfite conversion88.9
Moline 2013 [59]PCR55.9
Batte 2014 [148]Not described -
Bruegl 2014 [149]PCR-
Buchanan 2014 [69]PCR + bisulfite conversion
Goodfellow 2015 [71]Pyrosequencing and/or combined bisulfite restriction analysis70.3
Stelloo 2015 [75]PCR47.4
McConechy 2015 [77]PCR-
Goverde 2016 [150]Not described -
Watkins 2016 [79]PCR25.9
Lin 2016 [81]PCR64.7
Mills 2016 [82]PCR-
Ramalingam 2016 [83]PCR-
Shikama 2016 [84]PCR + bisulfite conversion-
Kato 2016 [85]Methylation-specific multiplex ligation-dependent probe amplification -
Okoye 2016 [86]PCR + bisulfite conversion75
Bruegl 2017 [151]PCR-
Zeimet 2017 [152]Not described -
Stelloo 2017 [90]PCR-
Dillon 2017 [91]PCR81.2
Najdawi 2017 [94]PCR-
Sloan 2017 [95]PCR15.8
Watkins 2017 [96]PCR + bisulfite conversion-
Adar 2018 [100]PCR + bisulfite conversion70
Pina 2018 [99]Not described 69
Kahn 2019 [112]Not described 53.9
Ryan 2019 [113]Not described 37.6
Sarode 2019 [116]Not described 26.7
Gan 2019 [122]PCR69.2
Ryan 2020 [124]NGS for germline mutation62.9
Rosa 2020 [125]PCR + bisulfite conversion + NGS for germline mutation48.8
Timmerman 2020 [127]NGS for germline mutation79
Missaoui 2020 [128]PCR + bisulfite conversion100
Dasgupta 2020 [129]Methylation-specific multiplex ligation-dependent probe amplification-
Ren 2020 [46]PCR + bisulfite conversion33.3
Rowe 2020 [136]PCR46.5
Stinton 2021 [137]Not described -
Tjalsma 2021 [139]Not described 14
Joehlin-Price 2021 [140]Not described 20
Yamamoto 2021 [141]PCR75
PCR: polymerase chain reaction.
Table 5. Prognosis of MMRd tumors compared with MMRp tumors in EC.
Table 5. Prognosis of MMRd tumors compared with MMRp tumors in EC.
ReferenceEC Total (n)Type of Tissue MMRd (n)* RFS or Recurrence MMRd* RFS or Recurrence MMRp** OS or Deaths MMRd** OS or DeathsMMRpPrognosis Conclusion
Parc 2000 [153]62Fresh + normal tissue21----Not significant
Ju 2006 [154]50FFPE12----Not significant
Choi 2008 [155]39FFPE8----Not significant
Shih 2011 [54]56FFPE95 year: 71.1%, 95% CI (53.1–89.1%)5 year: 97.6%, 95% CI (95.2–100%)5 year: 71.1%, 95% CI (53.1–89.1%)5 year: 100%MMRd was associated with worse RFS and OS compared with MMRp
Peiro 2013 [20]260FFPE33----Not significant
Ruiz 2014 [63]212FFPE64----Not significant
Stelloo 2015 [75]216FFPE195 year: 95%5 year: 93% POL-E, 52% no specific molecular profile 42% p53--MMRd was associated with a better RFS compared with MMRp
Mao 2015 [76]41Not described16----Not significant
Tangjitgamol 2017 [92]385FFPE2125 year: 67.0%, 95% CI (49.7–79.5%) advanced stage5 year: 40.0%, 95% CI (25.5–54.1%) advanced stage5 year: 66.5%, 95% CI (49.2–79.1) advanced stage5 year: 45.5%, 95% CI (30.2–59.5) advanced stageMMRd was associated with better RFS and OS in advanced stage compared with MMRp in advanced stage
Pina 2018 [99]892Not described242Recurrence: 10%Recurrence: 42%Deaths: 13.1%Deaths: 36.1%MMRd was associated with better RFS and OS compared with MMRp
Bosse 2018 [102]381Not described136HR = 0.61, 95% CI (0.37–1.00) compared with no specific molecular profileHR = 0.23, 95% CI (0.07–0.77) POLE compared with no specific molecular profileHR = 0.84, 95% CI (0.57–1.25) compared with no specific molecular profileHR = 0.56, 95% CI (0.27–1.15) POLE compared with no specific molecular profileMMRd was associated with better RFS and OS compared with MMRp
Backes 2019 [120]197Not described 645 year: 66%, 95% CI (45–79%)5 year: 89%, 95% CI (76–94%)5 year: 74%5 year: 86%MMRd was associated with worse RFS and OS compared with MMRp
Beinse 2020 [126]159FFPE35----Intermediate
Kolin 2020 [158]96FFPE3436 months9 months--MMRd was associated with better RFS compared with MMRp
León-Castillo 2020 [131]423FFPE1375 year: 71.7%5 year: 98% POL-E, 48% p535 year: 81.3%5 year: 98% POLE, 53% p53Intermediate
Kim 2020 [133]52FFPE5----Not significant
Joehlin-Price 2021 [140]95FFPE35----Not significant
Yamamoto 2021 [141]395FFPE68----Not significant
FFPE: formalin-fixed paraffin-embedded, MMRd: mismatch repair-deficient, MMRp: mismatch repair-proficient, RFS: recurrence-free survival, OS: overall survival. * RFS or recurrence is presented according to the data available; ** OS or death is presented according to the data available, HR: hazard ratio; POLE: polymerase-ε, CI: confidence interval.
Table 6. PDL-1 in MMRd tumors in endometrial cancer.
Table 6. PDL-1 in MMRd tumors in endometrial cancer.
ReferenceType of Tissue MMRd (n)MMRd (%)PDL-1
Jones 2020 [161]Not described20333PDL-1 was more frequent in MMRd tumors
Bregar 2017 [88]FFPE1333PDL-1 was present in 62% of MMRd tumors and high-grade tumors vs. 46% in MMRp tumors
Sloan 2017 [95]FFPE3856.7100% MMRd tumors demonstrated PDL-1 expression in peritumoral immune compartment
Rowe 2020 [136]FFPE4369.460.4% MMRd tumors showed positive tumoral PDL-1 vs. 5.3% MMRp
FFPE: formalin-fixed paraffin-embedded, MMRd: mismatch repair-deficient.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Favier, A.; Varinot, J.; Uzan, C.; Duval, A.; Brocheriou, I.; Canlorbe, G. The Role of Immunohistochemistry Markers in Endometrial Cancer with Mismatch Repair Deficiency: A Systematic Review. Cancers 2022, 14, 3783. https://doi.org/10.3390/cancers14153783

AMA Style

Favier A, Varinot J, Uzan C, Duval A, Brocheriou I, Canlorbe G. The Role of Immunohistochemistry Markers in Endometrial Cancer with Mismatch Repair Deficiency: A Systematic Review. Cancers. 2022; 14(15):3783. https://doi.org/10.3390/cancers14153783

Chicago/Turabian Style

Favier, Amelia, Justine Varinot, Catherine Uzan, Alex Duval, Isabelle Brocheriou, and Geoffroy Canlorbe. 2022. "The Role of Immunohistochemistry Markers in Endometrial Cancer with Mismatch Repair Deficiency: A Systematic Review" Cancers 14, no. 15: 3783. https://doi.org/10.3390/cancers14153783

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop